Ídia lexandra dos antos cunha - repositório … · lower atp synthesis, reduced mitochondrial...

139
LÍDIA ALEXANDRA DOS SANTOS CUNHA ACETYL-L-CARNITINE NEUROPROTECTION IN MITOCHONDRIAL DYSFUNCTION Tese de Candidatura ao Grau de Doutor em Patologia e Genética Molecular, submetida ao Instituto de Ciências Biomédicas Abel Salazar Universidade do Porto Orientadora: Doutora Maria Teresa Burnay Summavielle Categoria: Investigador Auxiliar Afiliação: Instituto de Biologia Molecular e Celular

Upload: ngokhue

Post on 03-Oct-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

LÍDIA ALEXANDRA DOS SANTOS CUNHA

ACETYL-L-CARNITINE NEUROPROTECTION IN MITOCHONDRIAL DYSFUNCTION

Tese de Candidatura ao Grau de Doutor em

Patologia e Genética Molecular, submetida ao

Instituto de Ciências Biomédicas Abel Salazar

Universidade do Porto

Orientadora: Doutora Maria Teresa Burnay Summavielle

Categoria: Investigador Auxiliar

Afiliação: Instituto de Biologia Molecular e Celular

Page 2: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 3: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

iii

DIRECTIVAS LEGAIS

Nesta Tese foram apresentados os resultados contido s nos artigos publicados ou

em vias de publicação seguidamente mencionados:

Cunha L , Horvath I, Ferreira S, Lemos J, Costa P, Vieira D, Veres DS, Szigeti K, Summavielle T,

Máthé D, Metello LF

Preclinical imaging: an essential ally in bioscienc es and drug development

(Submitted for publication)

Cunha L , Bravo J, Fernandes S, Magalhães A, Costa P, Metello LF, Horvath I, Borges F, Szigeti

K, Máthé D, Summavielle T

Acetyl-L-Carnitine preconditioning in methamphetami ne exposure leads to excessive

glucose uptake impairing reference memory and dereg ulated mitochondrial membrane

function

(Submitted for publication)

Summavielle T, Cunha L , Damiani D , Bravo J, Binienda Z, Koverech A , Virmani A (2011)

Neuroprotective action of acetyl-L-carnitine on met hamphetamine-induced dopamine

release . American Journal of Neuroprotection and Neuroregeneration 3:1-7.

Comunicações orais apresentadas em congressos inter nacionais:

Cunha L , Bravo J, Gonçalves R, Rodrigues C, Rodrigues A, Metello LF, Summavielle T

The role of mitochondria in acetyl-L-carnitine neur oprotective action

European Association of Nuclear Medicine Annual Congress. October 2012. Birmingham, UK.

Eur J Nucl Med Mol Imaging (2011) 38 (Suppl 2):S227

Page 4: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

iv

Apresentações sob a forma de poster em congressos i nternacionais:

Cunha L , Bravo J, Costa P, Fernandes S, Oliveira M, Castro R, Metello LF, Summavielle T

Acetyl-L-carnitine improves cell bioenergetics

European Association of Nuclear Medicine Annual Congress. October 2012. Milan, Italy.

Eur J Nucl Med Mol Imaging (2012) 39 (Suppl 2):S432

Summavielle T, Cunha L , Bravo J, Fernandes S, Costa P, Metello LF, Gonçalves R, Duarte M

Acetyl-L-carnitine neuroprotection and mitochondria bioenergetics

Society for Neurosciences Annual Congress. October 2012. New Orleans, USA

Cunha L, Damiani D, Alves C, Metello LF, Summavielle T

Study of mechanisms for acetyl-L-carnitine neuropro tective action

European Association of Nuclear Medicine Annual Congress. October 2010. Vienna, Austria.

Eur J Nucl Med Mol Imaging (2010) 37 (Suppl 2):S388

Apresentações sob a forma de poster em congressos n acionais:

Cunha L , Bravo J, Gonçalves R, Metello LF, Rodrigues A, Summavielle T

Mitochondrial function and acetyl-L-carnitine neuro protection

Congresso da Sociedade Portuguesa de Neurociências. Maio 2011. Lisboa, Portugal.

Bravo J, Cunha L , Fernandes S, Binienda Z, Summavielle T

Acetyl-L-carnitine neuroprotection through autophag y and UPS activity

Congresso da Sociedade Portuguesa de Neurociências. Maio 2011. Lisboa, Portugal.

Page 5: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

v

ABSTRACT

L-Carnitine (LC) is a naturally occurring compound responsible for the transport of long

chain fatty acids across the mitochondrial membranes into the matrix, where they undergo

β-oxidation. LC, absorbed from diet or endogenously synthesized, can be acetylated,

originating esterified compounds such as acetyl-L-carnitine (ALC). ALC plays a variety of

vital functions in the body, mainly related to the mitochondria. For instance, ALC is

involved in the transmitochondrial membrane trafficking of acetyl groups and,

consequently, in the control of acetyl-CoA/CoA intramitochondrial ratio, and therefore ALC

is also involved in the regulation of glucose oxidation. The close interaction between ALC

and cell bioenergetics, determines its involvement in many diseases, especially those

related to the mitochondria. ALC has been proposed to have beneficial effects in chronic

neurodegenerative disorders in which the main outcome from ALC supplementation has

been the improvement of mitochondrial function. Recently, our group has demonstrated

that pre-treatment with ALC confers effective neuroprotection against 3,4-

methylenedioximethamphetamine (MDMA)-induced neurotoxicity, preventing

mitochondrial oxidative damage and, most importantly, preventing the typical MDMA-

induced serotonin loss. Although the role of ALC at the mitochondrial level has been

widely investigated, the molecular mechanisms underlying the action of ALC remain

elusive. This dissertation aims to contribute for a better understanding of the

mitochondria-related molecular mechanisms underlying the action of ALC in brain

bioenergetics.

We used C57BL/6J mice to evaluate the role of ALC in brain glucose uptake. In addition,

methamphetamine (METH), known to cause hypoglycemia in the human brain, was used

to decrease glucose uptake and to clarify the role of ALC under glucose-deprived

conditions. Animals were divided into four groups and treated with: 1) saline (control), 2)

ALC (100 mg/kg), 3) METH (10 mg/kg) or 4) ALC + METH (30 min later). A radioactive

analogue of glucose molecule, 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG), was then

injected in the tail vein. In vivo images of the mice brain were acquired in a PET-MRI scan

for 50 minutes. Here, we observed a generalized increase in glucose uptake in several

brain regions immediately after the administration of METH when mice were

preconditioned with ALC, which was particularly evident in regions receiving a strong

dopaminergic input and therefore more susceptible to METH action, the striatum and the

prefrontal cortex.

Page 6: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

vi

Since carnitine supplementation activates the insulin growth factor 1 (IGF1) signaling

cascade and consequently promotes glucose utilization, we investigated if transcript levels

of Igf1 and Igf1 receptor were affected by the different experimental conditions. We found

that exposure to METH induced a significant decrease in Igf1 and Igf1R levels the

striatum. The combined administration of ALC and METH prevented the decrease in Igf1

levels. Of relevance, in this group, there was an augment in Igf1R in levels that paralleled

the increased glucose uptake. ALC by itself increased Igf1 expression, which may be

relevant for the putative neuroprotective features of the compound.

To further understand the mechanisms underlying ALC action, we used a dopaminergic

cell line (PC12 cells), exposed to METH in deprived glucose conditions (LG) to explore the

role of ALC in glucose uptake and mitochondrial function. Similarly to what was observed

in vivo, preconditioning with an ALC dose of 0.01 mM increased glucose uptake in PC12

cells exposed to METH in a LG medium. This was associated with decreased cell viability,

lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS,

decreased mitochondrial membrane potential and translocation of activated Bax to the

nucleus.

To understand the effect of ALC over the dopaminergic functionality, we exposed PC12

cells to METH and evaluated the intra- and extracellular levels of DA. Moreover, using a

radioactive compound, antagonist of dopamine D2 receptors (D2R), we acquired in vivo

images to evaluate the striatal binding ratio of the radiotracer. Our results revealed that, in

vitro, ALC can interfere with DA release, decreasing the amount of extracellular DA in the

presence of METH. These data corroborated those obtained in vivo, in which ALC was

able to counteract the effects of METH on D2R occupancy by increasing the receptor

displacement of METH.

Overall, our results show that ALC when combined with METH exerted its action by

increasing glucose uptake and Igf1R levels in the striatum. Concerning dopaminergic

functionality, ALC showed not only to increase the intracellular levels of DA, but also to

improve the availability of D2R by promoting METH displacement, relevant for its putative

neuroprotective features. In vitro data showed that the combination of ALC 0.01 mM and

METH increased glucose uptake at 24h. However this was associated to decreased

viability and ATP synthesis, increased ROS and loss of mitochondrial membrane

potential.

Page 7: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

vii

RESUMO

A L-carnitina (LC) é um composto natural, responsável pelo transporte dos ácidos gordos

de cadeia longa através das membranas mitocondriais até à matriz, onde decorre o

processo de β-oxidação. A LC, quer seja absorvida a partir da dieta, quer seja produzida

endogenamente, pode ser acetilada, originando compostos esterificados, como por

exemplo, a acetil-L-carnitina (ALC). A ALC desempenha diversas funções no organismo,

especialmente relacionadas com a mitocôndria. Por exemplo, a ALC está envolvida no

tráfego transmembranar de grupos acetil ao nível mitocondrial e, consequentemente, no

controlo da razão intramitochondrial de acetil-CoA/CoA, estando assim igualmente

envolvida na regulação da oxidação da glucose. Esta interacção entre a ALC e a

bioenergética celular é determinante no seu envolvimento em muitos processos

patológicos, particularmente os que estão relacionados com a mitocôndria.

Inclusivamente, a administração de ALC tem demonstrado benefícios ao nível das

patologias neurodegenerativas, cujo efeito principal se reflecte na melhoria da função

mitocondrial. Recentemente, o nosso grupo demonstrou que o pré-tratamento com ALC

confere neuroprotecção efectiva contra a toxicidade induzida pela 3,4-

metilenodioximetanfetamina (MDMA), prevenindo não só o dano mitocondrial oxidativo,

mas também a perda de neurónios serotoninérgicos, típica da acção da MDMA. Apesar

de o papel da ALC ao nível mitocondrial estar bem estudado, os mecanismos

moleculares subjacentes à sua acção não são totalmente compreendidos. Esta

dissertação pretende contribuir para uma melhor compreensão dos mecanismos

moleculares, relacionados com a mitocôndria, subjacentes à acção da ALC na

bioenergética cerebral.

Usamos ratinhos C57BL/6J para avaliar o papel da ALC na captação cerebral de glucose.

A metanfetamina (METH), conhecida por provocar hipoglicemia no cérebro humano, foi

utilizada com o objectivo de diminuir a captação da glicose e esclarecer o papel da ALC

em condições de carência de glucose. Os animais foram divididos em quatro grupos e

tratados com: 1) solução salina (controlo), 2) ALC (100 mg/kg), 3) METH (10 mg/kg), 4)

ALC + METH (30 minutos depois). Um análogo radioactivo da molécula de glucose, 2-

desoxi-2-[18F]fluor-D-glucose (18F-FDG), foi injectado na veia da cauda. Foram adquiridas

imagens in vivo dos cérebros dos ratinhos num tomógrafo PET-MRI durante 50 minutos.

Observámos um aumento generalizado na captação da glucose em várias regiões

cerebrais imediatamente após a administração de METH, quando os animais foram

precondicionados com ALC. Este aumento foi mais evidente em regiões como o estriado

Page 8: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

viii

e o córtex pré-frontal, que possuem um considerável input dopaminérgico e, portanto,

mais susceptíveis à acção da METH.

Dado que a suplementação com carnitina activa a cascata do factor de crescimento da

insulina 1 (IGF1) e, consequentemente promove a utilização de glucose, investigámos se

os níveis do gene Igf1 e do respectivo receptor (Igf1R) estariam afectados nas diferentes

condições experimentais. Verificámos que a exposição à METH induziu um decréscimo

significativo nos níveis de Igf1 e Igf1R no estriado. A administração conjunta de ALC e

METH preveniu o decréscimo nos níveis de Igf1. De salientar que neste grupo, se

verificou um aumento nos níveis de Igf1R, concomitante com um aumento na captação

da glucose. A ALC per si induziu um aumento dos níveis de Igf1, aspecto que poderá ser

relevante para o alegado efeito neuroprotector do composto.

No sentido de melhor compreender os mecanismos subjacentes à acção da ALC,

usámos uma linha celular dopaminérgica (células PC12), e expusemo-la à acção da

METH em condições de carência de glucose, explorando assim, o papel da ALC na

captação da glucose e na função mitocondrial. Semelhantemente ao que observámos in

vivo, o pré-tratamento com ALC aumentou a captação da glucose nas células PC12

expostas à METH, na presença de meio de cultura com baixa concentração de glucose.

Associado a este aumento na captação da glucose, verificámos uma redução da

viabilidade celular, da síntese de ATP e da função mitocondrial, bem como um aumento

dos níveis intracelulares de espécies reactivas de oxigénio (ROS), diminuição do

potencial de membrana mitocondrial, bem como a translocação da Bax activa para o

núcleo.

De modo a explorar o efeito da ALC na função dopaminérgica, expusemos as células

PC12 à METH e avaliamos os níveis intra- e extracelulares de dopamina (DA). Além

disso, utilizando um composto radioactivo, antagonista dos receptores D2 (D2R),

adquirimos imagens in vivo para avaliar a razão de ligação estriatal do radiotraçador. Os

nossos resultados revelaram que, in vitro, a ALC pode interferir com a libertação de DA,

diminuindo a quantidade de DA extracelular, na presença de METH. Estes dados estão

de acordo com os obtidos in vivo, nos quais a ALC foi capaz de contrariar os efeitos da

METH na ocupação dos D2R, promovendo a saída da METH dos mesmos.

Globalmente, os nossos resultados mostram que a ALC, quando administrada em

conjunto com a METH, exerce a sua acção promovendo um aumento da captação da

glucose e dos níveis de Igf1R no estriado. Relativamente à função dopaminérgica, a ALC

não só aumentou os níveis intracelulares de DA, como também melhorou a

Page 9: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

ix

disponibilidade dos D2R, promovendo a saída da METH, aspectos relevantes para as

características neuroprotectoras da ALC. Os dados adquiridos in vitro mostram que a

administração conjunta de ALC 0.01 mM e METH, aumentou a captação da glucose

pelas células PC12 às 24h. Contudo, este aumento foi acompanhado por uma diminuição

da viabilidade celular, da síntese de ATP e da perda do potencial de membrana

mitocondrial.

Page 10: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

x

Page 11: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xi

AGRADECIMENTOS

Uma importante meta da jornada que tem sido a frequência do Programa Doutoral em

Patologia e Genética Molecular, está prestes a ser atingida: a entrega do manuscrito.

Outra meta importante será a defesa pública do mesmo. Evidentemente, um trabalho

desta natureza nunca é o esforço de uma só pessoa. Reconhecendo esse facto, gostaria

de deixar uma palavra de gratidão a todos quantos me acompanharam no percurso e

que, com o seu contributo, me permitiram ultrapassar dificuldades e enriquecer o

presente trabalho, de tal modo que o alcance desta primeira meta se tornasse uma

realidade. O percorrer desta jornada foi muito importante não só para o meu

desenvolvimento pessoal, profissional e científico, como também me permitiu adquirir

novas competências em áreas distintas daquelas que estão relacionadas com a minha

formação de base. Sem falsas modéstias, tenho consciência que hoje sou uma

profissional diferente e com um leque de competências mais amplo. Acredito que isso já

se reflicta hoje e se tornará mais evidente no futuro do meu desempenho profissional,

pois de outra forma, todo o esforço (meu e daqueles que acreditaram em mim) terá sido

em vão…

Passo então a expressar a minha gratidão a todas as pessoas que, das mais diversas

formas, foram importantes para o desenvolvimento do presente trabalho:

A. Apoio profissional e/ou científico

Começo por endereçar o meu agradecimento à Professora Doutora Teresa Summavielle,

orientadora científica deste trabalho, que me acolheu, desde o primeiro momento, com

entusiasmo e boa vontade no Laboratório de Neuroprotecção do Instituto de Biologia

Molecular e Celular e que, com a sua experiência e os seus ensinamentos, me permitiu

desenvolver um espírito crítico face aos resultados obtidos. Agradeço igualmente a

dedicação, a disponibilidade e, acima de tudo, a confiança demonstradas.

À Dra. Joana Bravo, colega de laboratório e “camarada de luta”! Obrigada Joana pelo teu

auxílio ao longo destes anos, pela tua paciência e persistência e por, em cada dia e em

cada tarefa procurares fazer sempre melhor.

À Dra. Sílvia Fernandes, que se encontra a fazer um percurso semelhante ao meu,

obrigada pelo auxílio nas experiências. Espero que possas atingir a meta por que lutas o

mais brevemente possível!

Page 12: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xii

À Mestre Danira Damiani, cujo percurso do Mestrado se cruzou com o início da minha

jornada. Um agradecimento pelas longas horas de trabalho em conjunto, sempre com

boa disposição e competência.

Aos restantes investigadores do Laboratório de Neuroprotecção, agradeço a simpatia

com que me acolheram na equipa.

Aos meus superiores e colegas de trabalho, pelo seu incentivo sempre presente.

Neste apartado, um agradecimento muito especial ao Professor Luís F. Metello,

Coordenador do Curso de Medicina Nuclear da Escola Superior de Tecnologia da Saúde,

Instituto Politécnico do Porto (ESTSP.IPP), por sempre ter acompanhado de perto a

evolução deste trabalho, por toda a colaboração prestada na realização da parte prática e

pelo seu incentivo sempre presente. “Boss é boss!” … e este é daqueles não nos deixa

acomodar nem por um segundo, com a sua inquietude contagiante!

Ao Dr. Pedro Costa, colega de trabalho, agradeço o auxílio na realização dos testes de

captação da glucose (in vitro), bem como no processamento das imagens PET e SPECT.

Obrigada, Pedro pelo teu profissionalismo, pela tua dedicação e preocupação constantes.

Ao Dr. Domingos Vieira, igualmente colega de trabalho, agradeço a paciência, a

imaginação e a disponibilidade patentes no auxílio prestado na procura das melhores

soluções para o processamento das imagens de PET e SPECT.

A todos os colegas da Área Técnico-Científica de Medicina Nuclear, expresso a minha

gratidão pelo vosso incentivo permanente, por perdoarem algumas fases de menor

disponibilidade e por partilharem responsabilidades profissionais, mesmo quando isso

implicou um acréscimo de trabalho. Graças à vossa dedicação e empenho pessoal, em

conjunto fizemos importantes conquistas durante os últimos anos!

À Doutora Paula Sampaio, responsável do serviço “Advanced Light Microscopy” por me

ajudar a dar os primeiros passos na microscopia de fluorescência: “um admirável mundo

novo!” e pelas preciosas dicas.

À Doutora Raquel Gonçalves, investigadora do grupo “NEWTherapies” do Instituto de

Engenharia Biomédica (INEB), pela iniciação na citometria de fluxo.

À Doutora Margarida Duarte, investigadora de Parasitologia Molecular, pelos seus

conselhos úteis em termos de função mitocondrial.

Page 13: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xiii

I also would like to thank to Drs. Jan Breteling and Markus Diemling, respectively CEO

and Research Director from HERMES Medical Solutions for their permanent availability

and for the technical support on using advanced tools in imaging processing and for

always answering “present!” when we needed.

B. Apoio pessoal

Um agradecimento muito especial à minha família, pelo carinho, pelo suporte e

preocupação que desde sempre demonstraram. Aos meus pais, pelo seu exemplo de

vida, bem como pela educação que me proporcionaram e sem os quais, não seria quem

sou. Às minhas irmãs, com quem posso sempre contar, apesar da distância que por

vezes nos separa.

Ao Luís, meu companheiro de vida, sempre disposto a aliviar o meu fardo. Pelo privilégio

que tem sido partilhar a vida contigo, tu que és dono de uma boa disposição e

criatividade ímpares!

À minha amiga Ana Teles (quase Doutora!), cujo destino se cruzou com o meu logo no

início desta jornada. Muito obrigada pelo teu carinho e amizade!

C. Apoio institucional

Ao Instituto de Ciências Nucleares Aplicadas à Saúde (ICNAS), o meu agradecimento

pela cedência do 18F-FDG, que tornou possível a realização dos testes de captação da

glucose nas linhas celulares.

I also thank to the Semmelweis University for receiving me in the Nanobiotechnology & In

Vivo Imaging Center, allowing the performance of great experiences with the wonderful

equipment that allowed me to obtain those valuable PET, SPECT, CT and MRI images.

To Dr. Domokos Máthé, Director of CROmed Ltd, I thank for the opportunity and the

pleasure that was to work in the Department and to learn with his staff. Ildiko, thank you

for your sympathy and professionalism!

O meu agradecimento ao Instituto de Biologia Molecular e Celular, por ter permitido a

realização do presente trabalho no Laboratório de Neuroprotecção.

Page 14: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xiv

À Fundação para a Ciência e Tecnologia (FCT) pelo financiamento concedido do

Programa de Apoio à Formação Avançada de Docentes do Ensino Superior Politécnico

(Bolsa SFRH/PROTEC/49698/2009).

À Escola Superior de Tecnologia da Saúde do Porto e ao Instituto Politécnico do Porto

pelo apoio concedido através do Programa de Formação Avançada, materializado

através de apoio financeiro e redução de serviço docente. Este apoio foi de facto um

factor decisivo para o sucesso no atingimento desta meta. Por tudo isto e também por

ser, com toda a probabilidade, a instituição que mais directamente virá a beneficiar da

aquisição de conhecimentos e competências acrescidas que este trabalho permitiu

desenvolver, parece-me particularmente relevante deixar “duas palavras” a testemunhar

o meu apreço e mais sincero reconhecimento para todos os que estiveram envolvidos

nas tomadas de decisão inerentes a todas as etapas do processo de apoio ao Programa

de Formação Avançada de que beneficiei. Um bem-haja pela vossa Visão e sentido de

Missão para o Politécnico do Porto e para a ESTSP.IPP! Podem contar comigo para

demonstrar que este investimento e esta aposta foram bem aplicados!

Page 15: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xv

INDEX

CHAPTER 1

GENERAL INTRODUCTION ----------------------------------------------------------------------------------------------------------- 21

1.1. BRIEF HISTORY OF CARNITINE DISCOVERY -------------------------------------------------------------------------------------- 23

1.2. CARNITINE CHEMISTRY AND BIOCHEMISTRY ------------------------------------------------------------------------------------ 23

1.2.1. Exogenous Carnitine --------------------------------------------------------------------------------------------------- 25

1.2.2. Endogenous Carnitine ------------------------------------------------------------------------------------------------- 26

1.2.3. The Transport of Carnitine ------------------------------------------------------------------------------------------- 27

1.2.4. The Carnitine Pool ------------------------------------------------------------------------------------------------------ 30

1.2.5. Carnitine and Carnitine Esters -------------------------------------------------------------------------------------- 31

1.6. BIOLOGICAL FUNCTIONS OF CARNITINE AND MITOCHONDRIAL BIOENERGETICS --------------------------------------------- 33

1.7. MITOCHONDRIA AND NEURODEGENERATIVE DISEASES ------------------------------------------------------------------------ 44

1.8. METHAMPHETAMINE AS AN OXIDATIVE STRESS INDUCER --------------------------------------------------------------------- 56

1.9. CELLULAR AND ANIMAL MODELS ------------------------------------------------------------------------------------------------ 66

1.10. OBJECTIVES ---------------------------------------------------------------------------------------------------------------------- 69

CHAPTER 2

ACETYL-L-CARNITINE IN MITOCHONDRIAL BIOENERGETICS AND FUNCTIONALITY -------------------------------- 71

2.1. INTRODUCTION-------------------------------------------------------------------------------------------------------------------- 73

2.2. MATERIALS AND METHODS ------------------------------------------------------------------------------------------------------ 74

2.2.1. In Vivo Assessment of Glucose Uptake --------------------------------------------------------------------------- 74

2.2.2. Assessment of Brain Igf1 and Igf1R Transcript Levels by Real Time Polymerase Chain Reaction

(RT-PCR) --------------------------------------------------------------------------------------------------------------------------- 75

2.2.3. Cell culture and drug regimen --------------------------------------------------------------------------------------- 75

2.2.4. In Vitro Assessment of Glucose Uptake --------------------------------------------------------------------------- 76

2.2.5. Cell Viability Assay ----------------------------------------------------------------------------------------------------- 76

2.2.6. Determination of Mitochondrial Membrane Integrity -------------------------------------------------------- 77

2.2.7. Assessment of ATP Synthesis ---------------------------------------------------------------------------------------- 77

2.2.8. Assessment of Mitochondrial Mass-------------------------------------------------------------------------------- 77

2.2.9. Determination of Mitochondrial Membrane Potential (Δψm) --------------------------------------------- 78

2.2.10. Determination of Bax Activation and Location --------------------------------------------------------------- 78

2.2.11. Statistical Analysis ---------------------------------------------------------------------------------------------------- 79

2.3. RESULTS --------------------------------------------------------------------------------------------------------------------------- 80

2.3.1. ALC Preconditioning Increased Glucose Uptake in METH Exposed Animals ---------------------------- 80

2.3.2. Igf1 and Igf1R Regulation -------------------------------------------------------------------------------------------- 81

2.3.3. ALC Affects Glucose Uptake in a Dose-dependent Way ------------------------------------------------------ 83

2.3.4. ALC/METH Prevented the METH-induced Decrease in Mitochondrial Function ----------------------- 84

2.3.5. ALC/METH Decreased ATP Synthesis in Glucose Deprived Cells -------------------------------------------- 86

2.3.6. Increased ROS Formation Associated Translocation of Active Bax to the Nucleus -------------------- 88

2.4. DISCUSSION AND CONCLUSION -------------------------------------------------------------------------------------------------- 91

CHAPTER 3

NEUROPROTECTIVE ACTION OF ALC IN DOPAMINERGIC NEUROTRANSMISSION --------------------------------- 95

3.1. INTRODUCTION-------------------------------------------------------------------------------------------------------------------- 97

Page 16: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xvi

3.2. MATERIAL AND METHODS ------------------------------------------------------------------------------------------------------- 98

3.2.1. Cell Culture and Drug Regimen ------------------------------------------------------------------------------------- 98

3.2.2. Dopamine Quantification -------------------------------------------------------------------------------------------- 98

3.2.3. In Vivo Quantification of Dopamine Striatal Binding --------------------------------------------------------- 99

3.2.4. Statistical Analysis---------------------------------------------------------------------------------------------------- 100

3.3. RESULTS ------------------------------------------------------------------------------------------------------------------------- 101

3.3.1. ALC Action on Dopamine Release in PC12 Cells -------------------------------------------------------------- 101

3.3.2. ALC Neuroprotective Action over METH 1.0 µM Exposure ------------------------------------------------ 102

3.3.3. ALC Neuroprotective Action over METH 100 µM Exposure ------------------------------------------------ 103

3.3.4. ALC Neuroprotective Action over DA Receptors In Vivo ---------------------------------------------------- 105

3.4. DISCUSSION --------------------------------------------------------------------------------------------------------------------- 107

3.5. CONCLUSION -------------------------------------------------------------------------------------------------------------------- 112

CHAPTER 4

GENERAL DISCUSSION-------------------------------------------------------------------------------------------------------------- 115

REFERENCES --------------------------------------------------------------------------------------------------------------------------- 123

Page 17: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xvii

LIST OF FIGURES

Figure 1.1 – Chemical structure of L-carnitine ------------------------------------------------------------------- 24

Figure 1.2 – Schematic representation of LC biosynthesis --------------------------------------------------- 27

Figure 1.3 - Chemical structure of acetyl-L-carnitine ----------------------------------------------------------- 32

Figure 1.4 – Schematic representation of carnitine shuttle --------------------------------------------------- 35

Figure 1.5 – The mitochondrial spiral ------------------------------------------------------------------------------ 46

Figure 1.6 – Free radical production from catecholamines breakdown and autoxidation and glutamate receptors ---------------------------------------------------------------------------------------------------- 51

Figure 1.7 – Molecular structure of the most common amphetamines and its similarities with dopamine ----------------------------------------------------------------------------------------------------------------- 57

Figure 1.8 – Mechanisms through which METH increases synaptic levels of catecholamines ------ 58

Figure 1.9 – Temporal sequence of events associated to METH in vitro toxicity ----------------------- 60

Figure 1.10 – Positron emission tomography images of dopamine D2 receptors ---------------------- 63

Figure 1.11 – 18F-FDG PET images depicting brain glucose uptake under control condition or after METH administration -------------------------------------------------------------------------------------------------- 64

Figure 1.12 – Schematic representation of PC12 cells in comparison to dopaminergic nerve terminal -------------------------------------------------------------------------------------------------------------------- 67

Figure 2.1 - Increased glucose uptake in the brain after the administration of METH to mice preconditioned with ALC ---------------------------------------------------------------------------------------------- 81

Figure 2.2 - Igf1 (A) and Igf1R (B) transcript levels in the striatum of mice from different groups -- 82

Figure 2.3 - Altered glucose uptake in long time starvation in PC12 cells exposed ALC ------------- 84

Figure 2.4 – Cell viability of PC12 cells at 24 and 72h -------------------------------------------------------- 85

Figure 2.5 - Decreased mitochondrial function in conditions of long time starvation, induced by METH ---------------------------------------------------------------------------------------------------------------------- 86

Figure 2.6 – ATP synthesis of PC12 cells under different conditions -------------------------------------- 88

Figure 2.7 - Decreased membrane potential in cells preconditioned with ALC and exposed to METH ---------------------------------------------------------------------------------------------------------------------- 89

Figure 2.8 – Translocation of active Bax to the nucleus ------------------------------------------------------ 90

Figure 2.9 – The combined use of ALC and METH decreases mitochondrial mass ------------------- 91

Figure 3.1 – Effects of pre-treatment with ALC at 0.01, 0.1, 0.5, 1.0 mM on DA release ----------- 101

Figure 3.2 – Effects of pre-treatment with ALC at 0.01, 0.1, 0.5, 1.0 mM and METH 1.0 µM on DA release ------------------------------------------------------------------------------------------------------------------ 103

Figure 3.3 – Effects of pre-treatment with ALC at 0.01, 0.1, 0.5, 1.0 mM and METH 100 µM on DA release ------------------------------------------------------------------------------------------------------------------ 104

Figure 3.4 – ALC reverses the METH-induced increased D2 receptors occupancy ----------------- 106

Figure 3.5 – Representative 123I-IBZM SPECT/MR images from 12-week old male mice ---------- 107

Page 18: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xviii

LIST OF TABLES

Table 1.1 – Carnitine content of some foods -------------------------------------------------------------------- 26

Table 1.2 - Carnitine concentration in several organs --------------------------------------------------------- 30

Table 1.3 - Biological putative functions and effects of carnitine -------------------------------------------- 40

Table 1.4 – Main neurodegenerative diseases and mitochondria involvement ------------------------- 54

Table 1.5 – Effects of METH ----------------------------------------------------------------------------------------- 65

Page 19: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xix

LIST OF ABBREVIATIONS

123I-IBZM Iodine-123-labeled(S)-2-hydroxy-6-methoxy-N-((1-ethyl-2-pyrrolidinyl)methyl)

benzamide 18F-FDG Fluorine-18-labeled 2-deoxy-2-fluoro-D-glucose

3-NT 3-nitrotyrosine

5-HT Serotonin

AIF Apoptosis-inducing factor

ALC Acetyl-L-carnitine

AMPA α- amino-3-hydroxy-5-methyl-4-isoxazole propionic acid

ATP Adenosine triphosphate

Aβ β-amyloid

BBB Blood brain barrier

CAT Carnitine translocase

CoA Coenzyme A

COX Cytochrome-c oxidase

CPT I Carnitine palmitoyltransferase I

CPT II Carnitine palmitoyltransferase II

CuZn-SOD Copper-zinc superoxide dismutase

DA Dopamine

D1R Dopamine D1 receptor

D2R Dopamine D2 receptor

D3R Dopamine D3 receptor

DAT Dopamine transporter

DNA Deoxyribonucleic acid

DOPAC 3,4-dihydroxyphenylacetic acid

EHC Euglycemic hyperinsulinemic clamp

ETC Electron transport chain

FA Fatty acids

FADH2 Flavin adenine dinucleotide 2

FBS Fetal bovine serum

fMRI Functional magnetic resonance imaging

GLUT Glucose transporter

GSH Glutathione

GSH-Px Glutathione peroxidase

GTP Guanosine triphosphate

HG High glucose

HVA Homovanillic acid

IMM Inner mitochondrial membrane

Page 20: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

xx

KGDHC α-ketoglutarate dehydrogenase complex

LC L-carnitine

LG Low glucose

MAO Monoamine oxidase

MDMA 3,4-methylenedioximethamphetamine

METH Methamphetamine

MPT Mitochondrial permeability transition

MRI Magnetic resonance imaging

MRS Magnetic resonance spectroscopy

mtDNA Mitochondrial deoxyribonucleic acid

MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

NADH Nicotinamide adenine dinucleotide

NADPH Nicotinamide adenine dinucleotide phosphate-oxidase

NGF Nerve growth factor

NMDA N-methyl-D-aspartate

O2�- Superoxide radical

OMM Outer mitochondrial membrane

PD Parkinson’s disease

PDHC Pyruvate dehydrogenase complex

PET Positron emission tomography

PFC Prefrontal cortex

RNA Ribonucleic acid

RNS Reactive nitrogen species

ROI Region of interest

ROS Reactive oxygen species

RT-PCR Real Time Polymerase Chain Reaction

SNPC Substantia nigra pars compacta

SPECT Single photon emission computed tomography

SUV Standardized uptake value

TFAM Mitochondrial-transcription factor A

TH Tyrosine hydroxylase

VMAT Vesicular monoamine transporter

VTA Ventral tegmental area

Page 21: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

CHAPTER 1

GENERAL INTRODUCTION

Page 22: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 23: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

23

1.1. BRIEF HISTORY OF CARNITINE DISCOVERY

Carnitine (β-hydroxy-γ-N-trimethylaminobutyric acid) is a naturally occurring compound in

all mammalian species having important roles in the transport of fatty acids across the

inner mitochondrial membrane into the matrix where they undergo β-oxidation, a key

source of energy production (Evans and Fornasini, 2003; Sharma and Black, 2009).

Carnitine was first described in the beginning of the last century, in 1905, as a constituent

of muscle tissue, owing its name to the high concentration present in meat (from the Latin

word Carnis) (Gulewitsch and Krimberg, 1905). The chemical structure was only

established 22 years later (Tomita and Sendju, 1927). Despite intense research during the

1930s, stimulated by the similarities between carnitine and choline, it was not before the

1950s that its biological functions started to be unveiled, when researchers found an

essential growth factor for the mealworm Tenebrio molitor (common as fish food), called

vitamin BT (T from Tenebrio), that later on was identified as being L-carnitine (LC).

Subsequent studies showed that mealworm larvae growing in a state of carnitine

deficiency accumulated high amounts of fat, but still apparently died from starvation, as

they were unable to use their fat stores to survive. This suggested that LC might play a

role in fat oxidation. In the middle 1950s, two important papers renewed the interest on

carnitines and revealed some of the biological functions of carnitine in mammals. It was

shown that carnitine stimulates fatty acid oxidation in liver homogenates as well as that it

can be acetylated by acetyl coenzyme A (acetyl-CoA) in reversible way (Friedman and

Fraenkel, 1955; Fritz, 1955), leading to the discovery that carnitine functions as a carrier

of active acetyl groups through the mitochondrial membrane (Bremer, 1962). However,

these observations only acquired true clinical significance when the first cases of human

inborn errors in carnitine transport and metabolism were described, in 1973 (DiMauro and

DiMauro, 1973; Engel and Angelini, 1973). Since then, carnitine has become an

interesting topic of research over a variety of fields, namely, clinical medicine (neurology,

cardiology and oncology), sports medicine, nutrition and aging. A demonstration of this is

the number of papers on the subject listed in the “Pubmed” over the last ten years (above

4.300).

1.2. CARNITINE CHEMISTRY AND BIOCHEMISTRY

L-Carnitine (molecular formula: C7H15NO3; molecular weight: 161.2 g/mol) is a white,

crystalline and hygroscopic powder. As it is a zwitterion (dipolar salt) (figure 1.1), in

aqueous solution, LC is freely soluble in water since more than 90% of its ionizable

Page 24: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

24

groups (COO- and N+(CH3)3) dissociate at a physiological pH (~7.4) (Virmani and

Binienda, 2004).

Figure 1.1 – Chemical structure of L-carnitine (*binding sites for acyl residues).

The hydroxyl group at the C2 position (figure 1.1) is virtually undissociated in solution,

being of great physiological significance because it determines some of LC functions. LC

participates in reversible transesterification reactions, in which an acyl group is transferred

from coenzyme A to the hydroxyl group of LC, originating carnitine esters such as acetyl-

L-carnitine (ALC) and propionyl-L-carnitine (PLC). Other carnitine esters are also

biosynthesized in this manner (Rebouche, 1992; Sharma and Black, 2009). Due to its four

different ligands, the C-atom of the hydroxyl group is optically active and occurs in two

stereoisomeric forms, the D- and the L-form. Despite only the L-isomer is biological active,

the D-isomer is capable of interfering with the membrane transport of LC (Gross et al.,

1986; Wu et al., 1999).

LC is present in all animal species as well as in many microorganisms and plants

(although in the latter, the availability is limited). Carnitine homeostasis is maintained

through a relatively low rate of several mechanisms such as endogenous synthesis (25%),

absorption from diet (75%) through enterocyte membranes, renal reabsorption and other

mechanisms present in tissues that are able to maintain adequate concentration gradients

between intracellular and extracellular carnitine pools1 (Rebouche and Seim, 1998; Evans

and Fornasini, 2003; Virmani and Binienda, 2004; Flanagan et al., 2010). The status of

carnitine in humans varies with body composition, gender and diet. A brief description of

both exogenous and endogenous ways of carnitine provision will follow.

1 The term “carnitine pool” comprises free carnitine and short-, medium- and long chain carnitine esters, generally designated as acylcarnitines.

*

Page 25: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

25

1.2.1. Exogenous Carnitine

Exogenous carnitine may be absorbed from food or food supplements through active or

passive mechanisms and its bioavailability varies with diet composition (Rebouche and

Chenard, 1991; Evans and Fornasini, 2003). In vegetarians, who are used to low carnitine

diets, the bioavailability is superior (66% to 86% of available carnitine) comparatively to

regular red meat eaters, used to high carnitine diets (54% to 72% of available carnitine).

Omnivorous ingest 2-12 µmol/kg of body weight/day (23-135 mg) which represents 75%

of body carnitine sources. Bioavailability of oral carnitine dietary supplements is very low

comparatively to those obtained from food (14 to 18% of dose) and unabsorbed LC is

mostly degraded by microorganisms in the large intestine (Rebouche and Chenard, 1991;

Evans and Fornasini, 2003; Brass, 2004; Flanagan et al., 2010). On the other hand, orally

administered ALC is deacetylated soon after its uptake by the intestinal cells and part of

the recently formed free carnitine is the reacetylated (Gross et al., 1986). However,

although high doses of LC or ALC are generally well tolerated, the absorption of oral

dosage forms is very low (Rosca et al., 2009), as described by Parnetti et al. who treated

patients with senile dementia with 2.0 g/day for 50 consecutive days of oral ALC and

observed only a modest increase in plasma concentration (Parnetti et al., 1992). They

also observed that intravenous administration of 500 mg led to a rapid increase in plasma

concentration, with gradual clearance until reaching the baseline 12h after administration.

The most important metabolites are trimethylamine oxide in urine and γ-butyrobetaine in

feces (Rebouche and Chenard, 1991). Diet effects on plasma or serum free and total

carnitine concentrations among individuals of different gender and age has been

extensively studied and was summarized by Rebouche and Chenard (Rebouche and

Chenard, 1991). There is evidence that meat is the main source of LC. Dairy products,

seafood and fish are relatively low in carnitine whereas vegetables are generally very low

in carnitine (Demarquoy et al., 2004; Steiber et al., 2004). There is no recommended

dietary allowance or dietary reference intake for LC. Table 1.1 summarizes the carnitine

content of some selected foods.

Page 26: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

26

Table 1.1 – Carnitine content of some fo ods

Carnitine

(µmol/100 g) Carnitine

(mg/100 g)

Beef 401.0 65.0

Chicken 64.0 10.4

Turkey 131.0 21.2

Shrimp 4.0 0.7

Salmon 36.0 5.8

Milk dry whole 62.0 10.0

Milk 4% fat 14.0 2.3

Yogurt (regular) 75.0 12.2

Egg (white) 2.0 0.3

Egg (yolk) 5.0 0.8

Apple / Banana 1.0 0.2

Avocado 50.0 8.1

Aspargus / Spinach / Tomato 0.0 0.0

Potato 15.0 2.4

Garlic 8.0 1.3

Unabsorbed LC in the gastrointestinal tract is metabolized by bacteria of the intestinal

flora, originating trimethylamine and γ-butyrobetaine. Bacteria metabolism increases as

the oral intake of LC also increases, because in this case, the amount of LC that reaches

the gastrointestinal tract, available for breaking down by bacteria is higher (Evans and

Fornasini, 2003).

1.2.2. Endogenous Carnitine

Carnitine not obtained from diet, is endogenously synthesized from two essential amino

acids, lysine and methionine, primarily in the liver and kidneys. L-lysine provides the

carbon chain and nitrogen atom and L-methionine provides the methyl groups of LC

(Rebouche, 1992; Rebouche and Seim, 1998; Vaz et al., 1999; Virmani and Binienda,

2004). Synthesis of carnitine begins with methylation of lysine residues (in proteins like

myosin, actin and histones) by S-adenosylmethionine, the donor of methyl groups. 6-N-

trimethyl lysine is the first product of the reaction, being initially bound in proteins, is then

released through proteolysis in lysosomes. The next step occurs in the cytosol, where the

hydroxylation of 6-N-trimethyl lysine (C3) originates 3-hydroxy-6-N-trimethyl lysine. Then,

the cleavage of hydroxytrimethyl lysine originates glycine and γ-trimethyl-

aminobutyraldehyde. Oxidation of the carboxyl group results in the production of γ-

[Adapted from (Demarquoy et al., 2004)]

Page 27: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

27

butyrobetaine (deoxy L-carnitine), which is finally oxidized, leading to the formation of L-

carnitine (β-hydroxy-γ-N-trimethylaminobutyric acid) (Bremer, 1983; Rebouche, 1992;

Rebouche and Seim, 1998; Vaz et al., 1999). In humans, the major sites of carnitine

synthesis are the liver, the kidneys and the brain because these are the only tissues with

a high activity of γ-butyrobetaine dioxygenase (BBD), the enzyme that hydroxylates γ-

butyrobetaine to form carnitine (Ringseis et al., 2012). Other substances such as

magnesium, vitamins C, B3 and B6, iron and α-ketoglutarate, as well as the cofactors

required to originate S-adenosylmethionine (methionine, folic acid, vitamin B12, and

betaine) are vital participants in the endogenous synthesis of carnitine. Under normal

circumstances, the rate of LC synthesis is approximately 1-2 µmol/kg body weight/day,

being regulated by the availability of 6-N-trimethyl lysine. Thus, conditions that increase

protein methylation and/or protein turnover may increase the rate of LC biosynthesis

(Rebouche, 1992; Vaz et al., 1999; Steiber et al., 2004; Sharma and Black, 2009;

Flanagan et al., 2010). A scheme of the described process is shown in figure 1.2.

Figure 1.2 – Schematic representation of LC biosynt hesis

In humans, the enzyme that catalyzes the final step of LC synthesis, is present in liver,

kidneys and brain, meaning that inter-organ transport must occur in order to widely

distribute LC, making it available for all body tissues (Bremer, 1983; Flanagan et al.,

2010).

1.2.3. The Transport of Carnitine

L-carnitine and acylcarnitines are present in all tissues. In most of them, because the

concentration of carnitine is about 10 fold higher in tissues than in plasma, an active

Page 28: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

28

uptake of carnitine occurs. Plasma concentration of free carnitine is the result of a

dynamic balance with acylcarnitines (the ratio acylcarnitines/free carnitine ≤ 0.4 is

considered normal) (Bellinghieri et al., 2003). Circulating LC concentrations are

maintained at a fairly constant level of around 50 µM, predominantly through

efficient reabsorption by the kidneys (in humans, 90–98% of filtered carnitine is

reabsorbed under normal homeostatic conditions) (Rebouche and Seim, 1998; Evans and

Fornasini, 2003). The uptake rate is highly variable among different organs.

Concentrations of carnitine and acylcarnitines change as a function of dietary conditions.

During starvation and after a high-fat meal intake, the fraction of acetylated carnitine in the

liver and kidneys rises significantly, whereas a carbohydrate-rich diet lowers the levels of

acetylated carnitine in the liver (Jones et al., 2010). In humans, the decrease in plasma

LC does not occur immediately, but the increase in long- and short-chain acylcarnitines

during fasting or diabetic ketosis seem to be fast (Frohlich et al., 1978; Genuth and

Hoppel, 1979; Rebouche and Seim, 1998). Although plasma levels have no direct

correlation with brain levels, a remarkable increase in brain acylcarnitine concentration

was observed by Murakami et al. in neonatal starved rats comparing to control group, with

almost all of the increase attributed to short-chain acylcarnitines. They suggested that

during starvation, carnitine and its esters may be redistributed to the brain, which may use

them for energy supply through fatty acid oxidation or for the delivery of acetyl groups

(Murakami et al., 1997).

Carnitine reabsorption is also sensitive to the amount in the diet. For instance, if filtration

of LC increases as a result of higher consumption or after intravenous infusion, the

efficiency of reabsorption drops promptly (Rebouche and Seim, 1998). It has been shown

that LC as well as ALC and γ-butyrobetaine are synthesized in the kidneys, which are

then secreted into the tubular lumen. Once the kinetics of these metabolites by the

transport systems (mentioned below) are the same, the fraction excreted in the urine is

the result of both the amount present in the glomerular filtrate and the amount secreted in

the tubular lumen by the epithelial cells. A rapid intracellular synthesis of acylcarnitine

(from LC) or direct accumulation from the circulation, will lead to a higher secretion and,

consequently, to a higher proportion of acetylated species in the urine comparatively to

that in the circulation. In fact, kidneys play an important role in the regulation and

homeostasis of acylcarnitines (Rebouche and Seim, 1998). Only in case plasma carnitine

concentration exceeds the normal range (supraphysiologic levels), the excess is rapidly

eliminated as a consequence of saturation of the reabsorption mechanism (Ringseis et al.,

2012).

Page 29: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

29

The carnitine system consists of carrier proteins that transport carnitine across the

membranes and enzymes, carnitine acyltransferases that catalyze the reversible

equilibrium: acyl-CoA + L-carnitine � CoA + acyl-L-carnitine (Ramsay et al., 2001; Evans

and Fornasini, 2003; Sharma and Black, 2009). Carnitine transport is mediated by a family

of organic cation transporters (OCT), playing a pivotal role in the homeostasis of

intracellular carnitine. The most important members of this family are OCTN1, OCTN2 and

OCTN3. OCTN1 has a very low affinity for carnitine transport (Nalecz et al., 2004).

OCTN2 is the most important controlling factor for carnitine pools in the plasma

membrane (and needs sodium for the co-transport of carnitine, but not for other organic

cations) (Indiveri et al., 2010). OCTN3 is present in the peroxisomal membrane of

mammals, having the highest specificity for carnitine (Ramsay et al., 2001; Nalecz et al.,

2004; Sharma and Black, 2009). Carnitine and acylcarnitine accumulate in most tissues

through the high affinity, sodium-dependent organic cation transporter OCTN2, being

highly expressed in the heart, placenta, skeletal muscle, pancreas, kidneys, testis and

epididymis and poorly expressed in the brain, lungs and liver (Nalecz et al., 2004; Rytting

and Audus, 2005; Sharma and Black, 2009). Mutations in the OCTN2 gene can cause

primary systemic carnitine deficiency, markedly reducing serum carnitine levels (0–5

µmol/L) because most of the filtered carnitine will be eliminated through urine (Scaglia et

al., 1999). The discovery of OCTN2 as the responsible protein for carnitine transport was

presented by Nezu et al., who were the first to demonstrate that patients with systemic

carnitine deficiency have a loss of OCTN2 carnitine transporter function (Nezu et al.,

1999).

It has been reported that carnitine transport may also occur through a Na+-, Cl--dependent

way by an amino acid transporter ATB0,+ (Nakanishi et al., 2001), characterized as a low-

affinity transporter of carnitine, being expressed in the hippocampus, trachea, lungs,

mammary glands and intestinal tract (Sloan and Mager, 1999; Nakanishi et al., 2001;

Nalecz et al., 2004). In vitro studies demonstrated that both OCTN2 and ATB0,+ may be

involved in the transport of carnitine into the brain as they are expressed in the apical as

well as in the basolateral membrane of the blood brain barrier (BBB) (Kido et al., 2001;

Berezowski et al., 2004; Nalecz et al., 2004).

The Michaelis-Menten (Km) values mentioned in the literature for the tissue uptake of LC

are highly variable. In part, this variability is due to the method chosen to perform the

assay (Evans and Fornasini, 2003). Yet, it seems that brain and liver have Km values

considerably higher (1–5 mmol/L) than kidney (0.1–0.5 mmol/L) and cardiac and skeletal

muscle (20–100 µmol/L). However, direct comparisons between tissues and experimental

Page 30: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

30

conditions are difficult to establish, not only due to the presence of multiple transporters

with different Km and maximum rates (Vmax) values, but also to the presence of a passive

transport component (Bremer, 1983; Evans and Fornasini, 2003; Stephens et al., 2007).

1.2.4. The Carnitine Pool

Not only the Km values, but also the concentration of LC varies widely among different

tissues. Tissue distribution and uptake are partially controlled by hormones, especially in

the liver and epididymis (Bremer, 1983). The latter exhibits extremely high concentrations

of carnitine, which, in rats can reach 2000 times the plasma concentration. Skeletal

muscle is the second in the ranking, concentrating more than almost 4 mmol/kg,

representing 100 fold of plasma concentration. Brain, liver and kidneys contain

intermediate levels of 300–1000 µmol/kg. Plasma and extracellular fluid show very low

amounts (40-60 µmol/kg) of LC (Bremer, 1983; Harper et al., 1993; Evans and Fornasini,

2003). Table 1.2 summarizes the carnitine concentrations in some human tissues.

Table 1.2 - Carnitine concentration in several organs

Carnitine Concentration

(µmol/kg)

Skeletal muscle 1140 – 3940

Heart 610 – 1300

Liver 500 – 1000

Brain 500 – 1000

Kidney 330 – 600

Plasma 40 – 60

As can be seen, the ratios of tissue-to-plasma levels are remarkably high, emphasizing

the importance of maintaining adequate levels of the compound within the primary

sites of fatty acid oxidation (Evans and Fornasini, 2003). It has been estimated that the

total LC pool in a healthy standard (70 kg) adult is about 128 mmol (21 grams). Note that

measurements of LC plasma levels may not give reliable information about the real

carnitine status in the body, because this compartment accounts with less than 1% of the

total carnitine pool, making quantification difficult. Plasma levels of the most abundant

carnitine ester, ALC, are around 3-6 µmol/kg, meaning that the concentration of total

carnitine (including LC and ALC) is about 50–60 µmol/kg. LC and ALC do not bind to

[Adapted from (Angelini et al., 1992; Harper et al., 1993; Evans and Fornasini, 2003)]

Page 31: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

31

plasma proteins and although it is known that blood cells contain LC, binding to

erythrocytes seems to be minor or negligible (Evans and Fornasini, 2003). Rebouche and

Engel have conducted a pioneer study on the pharmacokinetics of an exogenous dose of

radiolabeled of L-[methyl-3H]-carnitine (Rebouche and Engel, 1984). The compound was

administered by intravenous injection to healthy volunteers as well as to patients with

carnitine deficiency pathologies. Blood samples were collected for up to 28 days. Results

identified three distinct compartments for carnitine in the body: the extracellular fluid

(representing the initial distribution volume), fast equilibrating tissues (most probable to be

the liver and the kidneys) and slow equilibrating tissues (cardiac and skeletal muscle). In

healthy volunteers, the mean residence times (turnover) of LC in these compartments

were approximately 1h, 12h and 191h, respectively, being the whole body turnover time

about 66 days. In fact, LC exhibits both a slow uptake and a prolonged residence time in

muscle tissue, reason why muscle LC content changes very little after acute or short-term

administration to healthy individuals (Rebouche and Engel, 1984). However, it has been

documented that chronic administration of either intravenous or oral forms of LC for a

sufficient length of time, increases muscle LC levels (Sharma and Black, 2009).

1.2.5. Carnitine and Carnitine Esters

L-carnitine is present in the body in both non-esterified and esterified forms. Short-chain

(C2 – C5) organic acids and medium- (C6 – C12) or long-chain (C14-C24) fatty acids are

transferred to and from CoA and the hydroxyl group of carnitine. These reversible

reactions are catalyzed by a group of enzymes designated as carnitine acyltransferases.

Acylcarnitine esters are produced intracellularly during the normal cell metabolic activity,

in conjunction with the above mentioned carnitine functions, namely the generation of

long-chain acylcarnitine esters in order to transport the fatty acyl moieties into

mitochondria that then undergo β-oxidation. In fact, long-chain fatty acids can only enter

mitochondria as acylcarnitine esters. Short- and medium-chain acylcarnitine esters are

generated either in mitochondria and peroxisomes, partially as a way to remove organic

acids from these organelles as high-energy compounds (Rebouche and Seim, 1998). In

turn, acylcarnitine esters may also have diverse functions, either at cellular or organ level.

For example, benefits of PLC in the contractile function of heart and in the protection of

the ischemic heart from reperfusion injury, are widely studied (Broderick et al., 2000; Felix

et al., 2001; Lango et al., 2005; Vargiu et al., 2008; Mingorance et al., 2011). ALC (figure

1.3) is the most abundant carnitine ester and has a putative antioxidant effect in

mitochondria (Shigenaga et al., 1994) by increasing the endogenous pool of antioxidant

Page 32: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

32

glutathione (Aureli et al., 1999) and promoting the activity of antioxidant enzymes (El-

Awady el et al., 2010; Hao et al., 2011; Haorah et al., 2011). In fact, antioxidant properties

of ALC are still a matter of controversy, mainly because in vivo studies not always match

the findings of in vitro studies. Schinetti et al. explained antioxidant properties of ALC

based on its iron-chelating ability in vitro (Schinetti et al., 1987). This was partially

corroborated by Reznick et al., who administered carnitine derivatives to ischemia-

reperfused rat hearts and observed that none of the carnitine derivatives were able to

scavenge peroxyl or superoxide radicals. However, propionyl-L-carnitine and propionyl-D-

carnitine were the only derivatives able to suppress hydroxyl radical production in the

Fenton system, probably by chelating the iron required for the generation of hydroxyl

radicals (Reznick et al., 1992). In vitro, ALC has demonstrated to protect the electron

transport chain (ETC) from electron leakage and superoxide production (Shen et al.,

2008).

ALC also shown to improve impaired mitochondrial DNA expression in the brain and heart

of aged rats (Gadaleta et al., 1990), to improve cognitive function after ischemic injury and

ameliorate damage induced by traumatic spinal cord injury through improvement of

mitochondrial function (Zanelli et al., 2005; Kobayashi et al., 2010; Goo et al., 2012; Patel

et al., 2012; Zhang et al., 2012). ALC will be further discussed in this work.

Figure 1.3 - Chemical structure of acetyl-L-carniti ne

As previously mentioned, starvation induces an increase in acylcarnitine in plasma and

liver (Yamaguti et al., 1996). After feeding, a quick decrease in plasma is observed and is

concomitant with the increase in liver concentration. It was shown that during fasting,

short-chain acylcarnitine esters (particularly ALC) uptake by muscles parallels the release

of non-esterified carnitine. These findings suggest that, carnitine esters in plasma might

be contributing for the exchange of metabolic fuels from the liver to the muscle (Rebouche

and Seim, 1998). Likewise, during high-intensity exercise, there is a marked increase in

Page 33: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

33

short-chain acylcarnitine esters concentration in skeletal muscle paralleled with a

decrease in free carnitine levels. Low-intensity exercise does not change carnitine

metabolism (Hiatt et al., 1989; Rebouche and Seim, 1998; Stephens et al., 2007). The

study of carnitine metabolism during exercise has given evidence to support the concept

that carnitine and acetyltransferase enzyme have an important contribution for the

modulation of the acyl-CoA to free (non-esterified) CoA ratio during metabolic stress

(Rebouche and Seim, 1998).

1.6. BIOLOGICAL FUNCTIONS OF CARNITINE AND MITOCHONDRIAL

BIOENERGETICS

A variety of important biological functions in the body for carnitine, emphasized by

carnitine deficiency diseases, has been described. Two of these functions, and perhaps

the most important, are the catalytic function in the mitochondrial combustion of fatty acids

and the metabolic function as a buffer for the excess of acyl residues. For the catalytic

function, only tiny amounts of LC are needed. In the metabolic function, free LC is

esterified (mainly to ALC), involving larger amounts of LC in the process (Jones et al.,

2010). ALC exerts many of its biological actions through the effects of LC itself (described

above) as well as acetyl moieties. The latter, may acetylate –NH2 and –OH functional

groups in amino acids as lysine, serine, threonine, tyrosine and N-terminal amino acids in

peptides and proteins, eventually modifying their structure, function and turnover. It has

been described that ALC has a higher potential for transacetylation reactions, transferring

acetyl groups more easily than acetylcholine or the terminal phosphate of ATP molecule

(Pettegrew et al., 2000).

Carnitine is essential for the shuttle of long-chain fatty acids across the inner

mitochondrial membrane, whose rate of β-oxidation is also controlled by LC. Thus, LC

plays an important role in energy metabolism (Rebouche and Seim, 1998). If alterations in

carnitine levels occur, this will have repercussions on energy production by the

mitochondria. The entry of long-chain fatty acids, released by lipolysis, into mitochondria

involves several steps. The first of them consisting in the activation of the fatty acid to CoA

occurs in the cytosol and originates fatty acetyl-CoA, which, however, is not able to cross

the mitochondrial membrane unless it is transported by carnitine (Kerner and Hoppel,

2000). This transport system consists of three distinct proteins: carnitine

palmitoyltransferase I (CPT I), carnitine translocase (CAT) and carnitine

palmitoyltransferase II (CPT II). CTP I, present in the outer mitochondrial membrane

Page 34: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

34

(OMM), catalyzes the transfer of the fatty acid moiety from long-chain fatty acyl-CoA to

carnitine (esterification of carnitine with long-chain acyl-CoA to form long-chain

acylcarnitine) and this is the rate-limiting step for β-oxidation of fatty acids, because CPT I

is inhibited by malonyl-CoA. In fact, there is an inverse relationship between the rate of

fatty acid oxidation and tissue malonyl-CoA concentration (Kerner and Hoppel, 2000;

Stephens et al., 2007). In the second step, the reaction products, long-chain

acylcarnitines, are transported from the outer to the inner mitochondrial membrane. This is

catalyzed by CAT that allows the transfer of one long-chain acylcarnitine molecule into the

mitochondria and the release of one molecule of free LC or acylcarnitine out of the

mitochondria (Kerner and Hoppel, 2000; Stephens et al., 2007). CAT also participates in

other functions such as buffering the mitochondrial acetyl-CoA/free CoA ratio and shuttling

chain-shortened fatty acid oxidation products from the peroxisomes into the mitochondria

(Ramsay and Arduini, 1993; Kerner and Hoppel, 2000). Once inside the mitochondria,

long-chain acylcarnitine molecules are transferred to CoA (transesterification of

acylcarnitine back to free carnitine and long-chain acyl CoA) through CPT II (an enzyme

located on the matrix side of the inner mitochondrial membrane), originating long-chain

acyl-CoA that is finally ready to undergo β-oxidation in which successive oxidative

removal of two carbon units in the form of acetyl-CoA occurs, starting at the carbonyl end

of the fatty acid chain (for example, palmitoyl-CoA yields eight acetyl-CoA molecules)

(Pettegrew et al., 2000). Carnitine is then transferred back to the intermembrane space

via CAT in an exchange with an incoming acylcarnitine. Each molecule of acetyl-CoA

produced from β-oxidation of fatty acids, originates one molecule of flavin adenine

dinucleotide (FADH2) and one molecule of nicotinamide adenine dinucleotide (NADH).

The acetyl-CoA is then oxidized to CO2 in the tricarboxylic acid (TCA) cycle, producing

one guanosine triphosphate (GTP) (or one adenosine triphosphate, ATP) molecule, three

NADH molecules and one FADH2 molecule. FADH2 and NADH undergo oxidation in the

ETC with each NADH molecule generating three ATP molecules and each FADH2

molecule generating two ATP molecules. In summary, each acetyl-CoA molecule

generated by β-oxidation of fatty acids can produce a total of 17 ATP molecules

(Pettegrew et al., 2000).

The link between β-oxidation and the electron pathway within the mitochondria originates

chemical energy. On the other hand, carnitine released in the mitochondrial matrix can

return to the cytosol via CAT. Therefore, carnitine has a pivotal role in the mitochondrial

utilization of long-chain fatty acids for energy production (Angelini et al., 1992; Carter et

al., 1995; Kerner and Hoppel, 2000; Stephens et al., 2007), which, in many organs,

Page 35: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

35

represent a crucial energy source (for example, liver and muscle). Figure 1.4 depicts the

carnitine shuttle and the above mentioned interactions.

Lipid storage in most organs occurs only in small amounts, making energy production

dependent on continuous supply of fatty acids mainly from adipose tissue. Once within the

cell, free fatty acids bind to fatty acids binding proteins, which are abundant in the cytosol.

Then, the fate of fatty acids depends on the tissue and its metabolic needs. It can be

converted into triglycerides or membrane phospholipids or oxidized in the mitochondria for

energy production. However, whatever the fate, fatty acids have always to be activated

first (Kerner and Hoppel, 2000). The acetyl-CoA produced is oxidized to CO2 and energy

(plus free CoA) in the TCA cycle of all mitochondria. It may be temporarily converted into

acetyl-L-carnitine and in ketone bodies in the fasting liver. These molecules are exported

to the blood and are oxidized via acetyl-CoA in other cells. In other organs than the liver,

the mitochondrial oxidation of fatty acids is deeply related with the TCA cycle.

Figure 1.4 – Schematic representation of carnitine shuttle [From (Finn and Dice, 2006), with permission].

On the other hand, the control of fatty acid oxidation and ketogenesis takes place in the

carnitine dependent transfer of activated fatty acids into the mitochondria, at CPT I level.

A high rate of fatty acid oxidation is correlated with a high acylcarnitine/free carnitine ratio.

The complete oxidation of acylcarnitines to CO2 and water is the result of two interacting

cycles, the β-oxidation cycle and the TCA cycle: one turn of the β-oxidation cycle

Page 36: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

36

originates one acetyl-CoA molecule and one turn of the TCA cycle converts one acetyl-

CoA into one CO2 molecule. In muscle, the two cycles work like gearwheels as there is

practically no use of acetyl-CoA outside the TCA cycle. Consequently, if the TCA cycle is

blocked, fatty acid oxidation will be inhibited in muscle tissues. In liver things are different

because the ketogenic enzymes are able to convert acetyl-CoA in to ketone bodies

(Bremer, 1983).

Due to the reversible transesterification of the acyl-CoAs with carnitine and the ability of

acylcarnitine to cross the mitochondrial membrane, the intramitochondrial relationship

between acyl-CoA and free CoA has an impact on the extramitochondrial acylcarnitine to

free carnitine ratio. This ratio, very sensitive to changes in mitochondrial metabolism, is

considered normal around 0.25 and abnormal above 0.4, representing a state of carnitine

insufficiency indicating that more carnitine is needed to handle any increased need for the

production of acylcarnitines (Angelini et al., 1992; Carter et al., 1995).

It was found by several groups that following a few minutes of intense exercise, there is a

remarkable reduction on free carnitine content in skeletal muscle (from approximately

75% of the total muscle carnitine pool to 20%), being this reduction correlated to the

formation of ALC, which in turn is associated to the increase of muscle acetyl-CoA.

Stephens et al. suggested that rate the of acetyl-CoA formation from pyruvate oxidation,

and catalyzed by the pyruvate dehydrogenase complex (PDHC), is in excess

comparatively to its rate of utilization by the TCA cycle, meaning that the rate of

condensation with oxaloacetate is less than its rate of production, leading to accumulation

(Stephens et al., 2007). The PDHC is a mitochondrial multi-enzyme complex that

regulates the oxidation of glycolytic pyruvate, generating acetyl-CoA for metabolism by the

TCA cycle or fatty acid synthesis, according to the following reaction (Randle, 1998):

Pyruvate + CoA + NAD+ → acetyl-CoA + NADH.

In the presence of increased PDHC flux (as during intense exercise), carnitine buffers the

excess of acetyl groups, ensuring that a viable pool of free CoA is kept for the

continuation of the PDHC and TCA cycle reactions. Moreover, the accumulation of ALC

itself provides a pool of acetyl groups that are available for transacetylation back to acetyl-

CoA, to be used in the TCA cycle when needed (Stephens et al., 2007).

Carnitine is responsible for maintaining adequate concentrations of free and esterified

CoA, as it provides a way for the removal of poorly metabolized and potentially toxic acyl-

CoA out of mitochondria (Carter et al., 1995; Ringseis et al., 2012). In fact, it has been

demonstrated that intracellular acyl-CoA derivatives accumulation contributes to the

Page 37: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

37

development of insulin resistance in skeletal and heart muscle. In this context, oral

carnitine supplementation has demonstrated to be a useful tool for the treatment or

prevention of insulin resistance and type 2 diabetes mellitus (Mingorance et al., 2011). In

addition, removal of acetyl-CoA disinhibits pyruvate dehydrogenase, leading to a

decrease in lactate levels and an increase in glucose oxidation (Stephens et al., 2007).

The role of carnitine (free carnitine and ALC) in reducing intra-mitochondrial acetyl-CoA

levels results in a 10- to 20-fold decrease in the acetyl-CoA/CoA ratio. This mechanism is

thought to be the responsible for the enhancement of glucose utilization by healthy and

type 2 diabetic individuals (De Gaetano et al., 1999; Mingrone et al., 1999). In a study

conducted by Mingrone et al. this stimulating effect on glucose oxidation was not

observed in healthy control subjects, but only in type 2 diabetic patients, whose PDHC

impaired activity was restored to normal as a result of carnitine infusion. In healthy

subjects, after carnitine infusion, an increase in glucose uptake was observed, but instead

of being used as a fuel, was stored in the form of glycogen (Mingrone et al., 1999).

Similarly, in a study with healthy subjects, under euglycemic hyperinsulinemic Clamp

(EHC) conditions, Stephens et al. observed an increase in glucose uptake, but also an

inhibition of glucose oxidation at the level of PDHC (increased muscle glycogen), probably

due to an increase in fatty acid oxidation mediated by carnitine (Stephens et al., 2006b).

Similar results were obtained by other groups, demonstrating an augmentation of non-

oxidative glucose disposal as a result of carnitine administration in EHC conditions

(Ferrannini et al., 1988; Wyss et al., 1990). These findings indicate that carnitine may

have an important role in the modulation of PDHC activity and thus, exert an effect on

whole body glucose homeostasis (Ringseis et al., 2012).

Carnitine has another important role in the mitochondria as it is a reservoir for excess acyl

residues, generated by high rates of β-oxidation, in which the acyl residue is

transesterified from CoA to carnitine, liberating CoA for other cellular reactions

(Rebouche, 2005; Ringseis et al., 2012). The acylcarnitine thus formed may have several

destinations: it may remain in the site of production and used when needed or may leave

from the cell and be used by other cells or tissues or it may be finally excreted from the

body. This represents an advantage of carnitine and its esters that unlike CoA and its

esters are able to easily cross most membranes, facilitated by specific carriers and

transporters. Clearly, this function has important implications for cellular energy

metabolism (Rebouche, 2005). For example, carnitine facilitates the oxidation of glucose

in working hearts by relieving inhibition of PDHC by fatty acids (Broderick et al., 1992).

The mechanism involves removal of acetyl groups generated from fatty acid β-oxidation

by transesterification from acetyl-CoA to carnitine, liberating CoA to participate in the PDH

Page 38: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

38

reaction. Carnitine may also increase the rate of glucose production and oxidation

secondarily by facilitating utilization of long-chain acyl-CoA in the hypothalamus. In

addition, it has been shown that inhibition of CPT I and, consequently, the increase in

long-chain acyl-CoA concentration in the hypothalamus, induces anorexia and decreases

hepatic glucose production (Obici et al., 2003).

Furthermore, it is thought that ALC may be a form of energy storage for the cell, at least in

sperm and macrophages. Peroxisomes also contain carnitine acyltransferases and a β-

oxidation enzyme system, particularly active in the shortening of very long chain fatty

acids (Bremer, 1990). Carnitine acyltransferases from peroxisomes seem to be active in

the transfer of the shortened acyl-CoAs and the acetyl-CoA to the mitochondria for

complete oxidation. The carnitine acyltransferases of the mitochondria can catalyze the

formation of PLC and branched chain acylcarnitines from branched chain amino acids and

methylthiopropionylcarnitine from methionine. According to Bermer, their formation

apparently represents a security system to prevent acyl-CoA accumulation in the

mitochondria (Bremer, 1990). In addition, another function attributed to carnitine is the

stabilization of cell membranes and enhancement of calcium transport (Hülsmann et al.,

1985).

Besides the modulation of the PDHC by carnitine, some authors have come up with

alternative explanations for the improvement of glucose tolerance associated to carnitine

supplementation, involving the modulation of expression of glycolytic and gluconeogenic

enzymes. In type 2 diabetic patients, oral ALC has been shown to correct a common

abnormality in these patients consisting in an inappropriate shift in the substrate use from

carbohydrates to lipids (Ruggenenti et al., 2009). Similar effect were observed after

intravenous infusion of L-carnitine in insulin resistant subjects with type 2 diabetes

mellitus, which increased whole body glucose utilization and decreased plasma free fatty

acids (Capaldo et al., 1991). Taken together, these results suggest that carnitine acts as

an essential cofactor for β-oxidation of fatty acids, facilitating the transport of long-chain

fatty acids across the mitochondrial membrane in the form of acylcarnitine esters. Other

mechanisms involve the modulation of key enzymes implicated in glycolysis and

gluconeogenesis (Stephens et al., 2007; Ruggenenti et al., 2009).

In another study, using a mouse model of primary carnitine deficiency (juvenile visceral

steatosis, JVS mouse model) it was demonstrated that hepatic transcript levels of

glycolytic enzymes (glucokinase and pyruvate kinase) are reduced, while hepatic

transcript levels of the gluconeogenic enzymes (phosphoenolpyruvate carboxykinase,

PCK1) are increased. Carnitine administration reversed these alterations in gene

Page 39: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

39

expression to normal levels, demonstrating the contribution of carnitine in the modulation

of genes involved in glucose metabolism (Hotta et al., 1996). More recently, carnitine has

also been shown to be involved in several other aspects related to glucose metabolism,

namely the increase in the expression of genes involved in glucose transport (e.g.,

GLUT8), the conversion of glucose into glucose 6-phosphate and glycolysis and the

down-regulation of genes involved in gluconeogenesis in pig liver. This shows that

suppression of hepatic gluconeogenesis also contributes for the positive effect of carnitine

on glucose utilization (Keller et al., 2011) and that the cause of this inhibitory effect on

gluconeogenic enzymes resides in the improvement of insulin action (Mingrone et al.,

1999). DNA microarray analysis in the liver of pigs whose diet was supplemented with 500

mg/kg of oral carnitine for 21 days, exhibited alterations in the expression of several

genes involved in insulin signaling cascade (insulin receptor substrate-2,

phosphatidylinositol 3-kinase regulatory alpha subunit and receptor protein-tyrosine

kinase erbB-3 precursor) (Keller et al., 2011).

The observation that carnitine improves glucose disposal and oxidation in healthy

volunteers is indicative that carnitine influences glucose metabolism acting at insulin

receptor level, by increasing transmembrane glucose transport or at post-receptor level,

influencing the insulin signaling cascade (Ferrannini et al., 1988; De Gaetano et al., 1999).

More recently, it was found that carnitine also has beneficial effects through its action on

the regeneration of endocrine pancreas, namely through the modulation of the insulin-like

growth factors (IGFs) and IGF binding proteins (Heo et al., 2001; Molfino et al., 2010).

Molfino et al. found that caloric restriction increases intestinal uptake of carnitine, which,

associated to oral carnitine supplementation, improves insulin resistance and may be an

adjunctive treatment in patients with impaired fasting glucose and type 2 diabetes mellitus

(Molfino et al., 2010). It has been shown that carnitine administration to diabetic rats

increases plasma concentrations of IGF-1 and IGF-2, possibly activating the IGF-1

signaling pathway, improving glucose tolerance (Heo et al., 2001).

In cardiac disease, the administration of carnitine to patients has also shown beneficial

effects by improving heart function. Surprisingly, the improvement of heart function in

diabetic or carnitine-deficient rat hearts was not due to an improvement in fatty acid

oxidation rates but to an increase in the overall glucose utilization (Broderick et al., 1992;

Broderick et al., 1995b; Broderick et al., 1995a). The reason for this is that carnitine

increases cytosolic acetyl-CoA, which in turn increases malonyl-CoA. As stated before,

the latter is a CPT I potent inhibitor. Indirectly, carnitine is decreasing fatty acid oxidation,

by decreasing fatty acid entry into the mitochondria (Broderick et al., 1995b; Broderick et

Page 40: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

40

al., 1995a). On the other hand, the transfer of intramitochondrial acetyl groups from

acetyl-CoA to carnitine (forming ALC) relives PDHC inhibition, thus increasing glucose

oxidation (Broderick et al., 1992). The carnitine derivative PLC is able to improve both

fatty acid oxidation and glucose utilization in diabetic hearts, because the propionyl group

is able to enter the TCA cycle as succinyl-CoA stimulating the TCA cycle flux and

ultimately, stimulating glucose utilization. On the other hand, the carnitine moiety released

from this reaction can also enhance fatty acid oxidation through the stimulation of the

carnitine shuttle system, therefore, preventing the accumulation of lipid intermediates and

improving insulin sensitivity (Schonekess et al., 1995). Table 1.3 summarizes the main

known biological functions and effects of carnitine.

Table 1.3 - Biological putative functions and effec ts of carnitine

Function References

Promotion of mitochondrial oxidation of long-chain fatty acids

(Pande and Blanchaer, 1970; Kerner and

Hoppel, 2000; Jones et al., 2010; Ringseis

et al., 2012)

Stimulation of mitochondrial metabolism by decreasing acyl-CoA and

increasing free CoA

(Bremer, 1990; Broderick et al., 1992;

Sharma and Black, 2009)

Membranes stabilization, abolishment of Ca2+-overload and stimulation

of microcirculation in ischemia (Hülsmann et al., 1985; Aureli et al., 2000)

Involvement in membrane repair by reacylation (Arduini et al., 1994)

Stimulation of immune response and decreasing of inflammatory

cytokines (Famularo et al., 1994; Winter et al., 1995)

Promotion of the maturation process of the fetus, lungs and sperm (Lohninger et al., 1996; Palmero et al.,

2000; Kang et al., 2011)

Decreasing of oxidative stress (Aureli et al., 1999; El-Awady el et al., 2010;

Hao et al., 2011)

Inhibition of apoptosis and opening of the permeability transition pore.

Inhibition of caspases

(Pastorino et al., 1993; Cifone et al., 1997;

Mutomba et al., 2000)

Protection against (neuro)toxins and correction of mitochondrial, lipid

and neurotransmitter receptor abnormalities in aging brain and heart

(Gadaleta et al., 1990; Aureli et al., 1994;

Aureli et al., 2000; Mazzio et al., 2003a;

Virmani et al., 2003; Kobayashi et al., 2010)

Prevention of mitochondrial damage and ATP depletion (Dhitavat et al., 2005)

Enhancement of glucose utilization by stimulating the PDHC activity (Ferrannini et al., 1988; Uziel et al., 1988;

Wyss et al., 1990; Stephens et al., 2006a)

Improvement of mitochondrial bioenergetics following spinal cord injury (Patel et al., 2012)

Amelioration of arterial hypertension, insulin resistance and impaired

glucose tolerance in subjects at increased cardiovascular risk (Ruggenenti et al., 2009)

Improvement of learning capacity in rats (Kobayashi et al., 2010)

Modulation of glucose metabolism and stimulation of glycogen

synthesis (Aureli et al., 1998)

Page 41: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

41

Stephens et al. proposed that an increase in carnitine-mediated long-chain fatty acid

oxidation, might inhibit glycolytic flux and carbohydrate oxidation (decreasing the PDHC

activity and lactate content and increasing glycogen storage), even in the presence of high

carbohydrate content (Stephens et al., 2007). This is a process known since the 1960s

from studies conducted by Randle et al. on rat hearts and diaphragm muscle (Randle et

al., 1963; Randle et al., 1964). According to Randle’s glucose – fatty acid cycle, an

increase in β-oxidation will enhance acetyl-CoA concentration and, consequently, citrate

and glucose-6-phosphate content will increase. This, in turn, results in the down-

regulation of carbohydrate flux, due to product inhibition of PDHC, phosphofructokinase

and hexokinase, respectively (Randle, 1998). In fact, increases in the mitochondrial ratio

of acetyl-CoA/CoA will result in the activation of PDH kinase, resulting in an increased

phosphorylation and consequently in the inhibition of PDHC. This will result in a decrease

in glucose oxidation rates. An important determinant of the ratio of acetyl-CoA/CoA is both

the rate of removal of intramitochondrial acetyl-CoA by the TCA cycle and the rate of fatty

acid β-oxidation, which is an alternate source of acetyl-CoA (Randle cycle).

The above mentioned effects of free LC can also be applied to ALC, which after ingestion,

is in large part hydrolyzed to L-carnitine (half-life: 10 to 45 hours) (Rebouche, 2004;

Ruggenenti et al., 2009). The acetyl moiety is either rapidly used or stored as acyl-esters

of varying chain length, in particular, in skeletal and cardiac muscles (Rebouche, 2004).

ALC is the main acylcarnitine ester, both intracellularly and in the plasma. Like LC, ALC

participates in both anabolic and catabolic pathways in cellular metabolism. 14C from

[acetyl-1-14C] acetyl-L-carnitine injected into mice appeared both as 14CO2 and in the

phospholipid and triacylglycerol fractions predominantly in liver (Farrell et al., 1986;

Rebouche and Seim, 1998).

In the adult brain, about 80% of total carnitine remains in the free from, 10-15% is present

as ALC and about 10% as long-chain acylcarnitines. In the developing brain (as observed

in suckling rats), the amount of ALC is higher, while free from diminishes by 40% (Nalecz

et al., 2004). Due to the similarities in the chemical structure between ALC and

acetylcholine, it has been proposed that ALC might play a role in neurotransmission

(cholinomimetic activity) (Falchetto et al., 1971; Pettegrew et al., 2000). ALC, like free

carnitine, is both transported across the BBB through OCTN2 and synthesized in the brain

(Wawrzenczyk et al., 1995; Nalecz et al., 2004). It has been reported that ALC crosses

more easily cell membranes and the BBB compared to free carnitine (Rebouche, 1992).

Two substrates are needed for acetylcholine synthesis: acetyl-CoA and choline. Choline is

able to cross the BBB and may originate from the intracellular pool, while acetyl-CoA can

Page 42: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

42

be synthesized in peroxisomes as well as in mitochondria. Acetyl-CoA generated inside

the mitochondria can cross the inner mitochondrial membrane in the direction of cytosol,

as citrate or as ALC. However, ALC is the main source of acetyl moieties for acetylcholine

synthesis in the brain (Tucek, 1990; Rosca et al., 2009). In addition, it was demonstrated

that in case of acetylcholine content depletion, its synthesis could be stimulated by the

administration of glucose, carnitine or ALC (Tucek, 1983). Studies performed with cortical

neurons and neuroblastoma cell line, showed that the addition of carnitine increased the

production of acetylcholine in the presence of glucose and choline (Wawrzenczyk et al.,

1995).

ALC has also been shown to improve membrane cytoskeletal protein-protein interactions,

increasing membrane stability and possibly altering cell membrane dynamics and

modulating its function (Arduini et al., 1990). Other evidences that ALC induces changes

in membranes was found by Paradies et al., who observed that after ALC administration,

there was a recovery on mitochondrial membrane phospholipid composition (cardiolipin)

and a restoration of phosphate carrier activity, which were altered as a result of aging

(Paradies et al., 1992).

A putative role of ALC in nerve growth factor (NGF) levels has been reported. NGF

regulates neuronal development and maintenance of the differentiated state in certain

neurons of the peripheral and central nervous system of mammals. ALC shown not only to

improve NGF-binding capacity in the hippocampus and in the basal forebrain regions of

aged rats, improving cognitive performance (Angelucci et al., 1988; Taglialatela et al.,

1996), but also to increase NGF receptor expression in the striatum of developing rats (De

Simone et al., 1991).

As will be presented in the next section, mitochondria-related enzymes are involved in the

process of neurodegeneration. A considerable number of both in vivo and in vitro studies

demonstrated that ALC is able to modulate a variety of mitochondrial and other

intracellular enzymes. Examples are the increase in the activity of key enzymes such as

cytochrome oxidase and α-ketoglutarate dehydrogenase induced by ALC, particularly in

synaptic mitochondria and acetylcholinesterase in plasma membrane from rat frontal

cortex (Gorini et al., 1996; Gorini et al., 1998). According to the literature, ALC exerts its

actions possibly through direct physicochemical interaction or through protein acetylation

(Pettegrew et al., 2000).

Furthermore, ALC has been described as having neuromodulatory effects on synaptic

transmission, namely by restoring the number of synaptic vesicles of giant synapses in the

Page 43: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

43

hippocampus of aged rats (Laschi et al., 1990). ALC also increases choline uptake into

nerve terminals, increasing acetylcholine synthesis, and enhances acetylcholine release

in the striatum and hippocampus of freely moving rats (Imperato et al., 1989). ALC is able

to attenuate the age-related reduction of a number of central nervous system receptors,

including N-methyl-D-aspartate (NMDA), NGF and glucocorticoid receptors and prevents

NMDA receptor loss in the hippocampus, striatum and frontal cortex of aged rats

(Castorina et al., 1994).

Fariello et al. found that ALC administration to mice for 5 consecutive days raised nigral

GABA, but striatal levels of dopamine and metabolites were not significantly affected. The

authors suggested that ALC would be useful in treating symptoms of neuronal dysfunction

related to the accumulation of metabolic waste (Fariello et al., 1988). Intracerebral ALC

administration by microdialysis in different brain regions, increases the release of

neurotransmitters as dopamine and acetylcholine as well as amino acids, particularly

aspartate, glutamate and taurine in a concentration-dependent manner (Toth et al., 1993).

The repeated administration of ALC led to consistent increases in DA output in the

nucleus accumbens, subsequent to acute stress conditions exposure (Tolu et al., 2002).

More recently, Zaitone et al. observed increased striatal DA levels in rats treated with

rotenone (an inhibitor of mitochondrial complex I) and ALC. This increase was not

observed the in control group, suggesting that ALC increases DA synthesis and release

as a response to injury (Zaitone et al., 2011). Using 13C-ALC, it was demonstrated that the

acetyl moiety of ALC is used as an energy source by both astrocytes and neurons and

rapidly incorporated into glutamate and GABA synthesis (Scafidi et al., 2010b). This

clarified the direct involvement of ALC in increased glutamine, glutamate and GABA

levels, through α-ketoglutarate production, via the tricarboxilic acid (TCA) cycle.

Importantly, biosynthesis of both tyrosine and L-tryptophan, dopamine and 5-HT

precursors, involves either a transamination reaction with glutamate or the participation of

glutamine as an amine donor (Yudkoff, 1997), which may account to explain the action of

ALC over monoamine production.

Moreover, ALC has a putative neuroprotective effect, both in vitro and in vivo, against a

variety of drugs such as ethanol (Rump et al., 2010; Abdul Muneer et al., 2011a; Haorah

et al., 2011), 1-methyl-4-phenylpyridinium (MPP+) (Mazzio et al., 2003b; Virmani et al.,

2004), methamphetamine (METH) (Coccurello et al., 2007; Abdul Muneer et al., 2011b),

3,4-methylenedioximethamphetamine (MDMA) (Alves et al., 2009) and rotenone (Zaitone

et al., 2011). These ALC features, as well as its neuromodulatory ability will be further

discussed.

Page 44: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

44

Since the human body is able to endogenously synthetize carnitine in sufficient amounts,

normally it is not required in the human diet. Until now, no clinical pathological conditions

have been associated to carnitine nutritional deficiencies. Vegans, who normally absorbed

lower carnitine amounts from diet, have lower plasmatic amounts, but no relevant clinical

deficiency (Rebouche, 1992). However, in some conditions, carnitine deficiencies may

develop, meaning that carnitine concentration in plasma or tissues is under the required to

assure the normal function of the body. Clinically, carnitine deficiencies are diagnosed

based on plasma levels of carnitine (below 2 µmol/L) and may have genetic or acquired

origin (medical conditions or iatrogenic states) (Pons and De Vivo, 1995).

Primary systemic carnitine deficiency of genetic origin results in mutations in the gene for

carnitine transporter OCTN2, which causes poor intestinal absorption of dietary LC and

impaired LC reabsorption by the kidneys, increasing urinary loss of the compound. Clinical

manifestation of this deficiency occurs in early childhood and includes cardiomyopathy

and often skeletal myopathy and/or hepatic encephalopathy. There are criteria to establish

this condition, namely: decreased plasma or tissue carnitine levels, impaired fatty acid

oxidation, the metabolic abnormalities are corrected when carnitine concentration is

restored to normal levels, absence of other primary defects in fatty acid oxidation (Treem

et al., 1988; Pons and De Vivo, 1995). Without treatment, primary systemic carnitine

deficiency is fatal. Treatment consists of pharmacological doses of carnitine.

Secondary carnitine deficiencies are more common and include both genetic and acquired

conditions, characterized by decreased carnitine levels in plasma or tissues or both.

Hereditary causes include genetic defects in amino acid degradation and lipid metabolism

leading to a build-up of organic acids, which are subsequently removed from the body via

urinary excretion of acylcarnitine esters. Kidney malfunction also results in increased

urinary losses of carnitine, by decreasing renal reabsorption of carnitine. Moreover, the

administration of some drugs also induces carnitine deficiencies (e.g. anthracyclines,

valproate, pivaloyl-antibiotics, cephalosporins, salicylic acid and aspirin, etc.) (Pons and

De Vivo, 1995).

1.7. MITOCHONDRIA AND NEURODEGENERATIVE DISEASES

Mitochondria are double membrane organelles enclosing a matrix playing a central role in

cell bioenergetics and cell death pathways. These two membranes invaginate, forming

mitochondrial cristae. Mitochondria have their own DNA (mtDNA), composed by 16.500

Page 45: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

45

base pairs and 37 genes, being rather complex and dynamic organelles, as they are able

to move, fuse and split. Besides mtDNA, the mitochondrial matrix also contains RNA,

detoxification systems and enzymes involved in β-oxidation and TCA cycle (Filosto et al.,

2011). The outer membrane is permeable to ions and solutes up to 50 kDa (Harris and

Thompson, 2000). The intermembrane space contains proteins encoded by nuclear DNA,

such as apoptosis promoting proteins (Harris and Thompson, 2000) and metallic ions and

enzymes responsible for several metabolic processes (Terziyska et al., 2005).

The inner membrane is impregnated with protein complexes (I–IV) constituting the ETC

through which the reducing equivalents coming from TCA cycle and β-oxidation pass

along, as well as two electron carriers (coenzyme Q and cytochrome c). Energy

conservation is maintained through charge separation at the level of the inner

mitochondrial membrane. The oxidation of substrates gives rise to electrons that are

transported to oxygen by the redox carriers of the respiratory chain simultaneously with H+

ejection on the redox pumps at complexes I, III and IV. This creates an electrochemical

proton gradient, which at normal physiological conditions is equivalent to -220 mV

(Bernardi et al., 1999) and is used to generate ATP. As mitochondria are the main power

plants of cells, any condition that interferes with energy homeostasis may be translated

into pathological conditions. The degree of dependence of a certain organ from oxidative

phosphorylation will determine the number of mitochondria. Because they are huge

energy consumers, the brain, heart and skeletal muscle are quite abundant in

mitochondria. Beyond their well-known role in ATP synthesis, mitochondria are a source

of reactive oxygen species (ROS), being involved in cellular responses to oxidative stress

and apoptosis (Filosto et al., 2011).

In the recent years, increasing interest has arisen on the possible role of mitochondria in

the onset and development of neurodegenerative diseases as it participates in a diversity

of biochemical processes such as energy production and reactive oxygen species

generation, regulation of intracellular calcium (Ca2+), excitotoxicity and apoptotic cascade

(Filosto et al., 2011). The question of whether are the oxidative stress and mitochondria

impairment the responsible for the onset and development of neurodegenerative diseases

or are the former a consequent of the latter is still to be answered (Petrozzi et al., 2007;

Filosto et al., 2011). In fact, energy impairment in the brain is tightly correlated to

intracellular Ca2+ regulation as well as ROS generation (Blass, 2000). It has been reported

that impairments in glucose metabolism seems to impair the mechanisms that quench

free radicals, contributing to free radical damage. This idea has been formulated as “the

mitochondrial spiral”, described by Blass (figure 1.5) (Blass, 2000). Moreover,

abnormalities in glucose oxidation are associated to neurodegenerative diseases as

Alzheimer’s and Parkinson’s Diseases (Blass, 2000; Hu et al., 2000). The reduction in

Page 46: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

46

cerebral metabolic rate occur prior to functional impairments are evident on

neuropsychological testing or brain atrophy being detected by neuromorphological

imaging, but they can induce cognitive impairments in AD patients (Blass, 2000).

Beneficial effects as a result of glucose or insulin administration indicate that the defects

in glucose metabolism in AD brain are functionally significant (Boyt et al., 2000; Craft et

al., 2000).

A variety of factors may contribute to trigger the mitochondrial spiral in the brain, such as

vascular disease, glutamate excitotoxicity, mitochondrial or nuclear DNA mutations,

toxins, etc. Initiation of mitochondrial spiral has an early impact on cellular function as

energy charge decreases, biosynthetic and other anabolic processes are spared in order

to maintain cell structure and osmotic regulation. Mitochondrial spiral can also result in cell

death due to apoptotic (cytochrome c release) or necrotic mechanisms (loss of ion

homeostasis), or the combination of both (Blass, 2000).

Figure 1.5 – The mitochondrial spiral. Energy impairments, changes in calcium homeostasis and

excessive ROS production interact with in mitochondria. Inducing any one of them leads to

abnormalities in the other two. The arrows pointing to initiators of the mitochondrial spiral are

grouped only for graphic convenience. Initiators can start the cycle at several points (e.g. glutamate

excitotoxicity may work directly on dysregulating Ca2+ and reperfusion injury after ischemia

damage, directly increases ROS. [From (Blass, 2000), with permission]

The brain cortex is mainly constituted by neurons and astrocytes; other cellular types,

which account for only a small proportion of the brain, are oligodendrocytes, microglia and

nonparenchymal cells like brain endothelial cells. In humans, astrocytes represent about

Page 47: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

47

30% of brain cortical volume, exhibiting the correspondent fraction of brain glucose

oxidation (one-third). About two-thirds of glucose metabolism in cerebral cortex occurs in

neurons. Oligodendrocytes, prominent in the white matter, exhibit low levels of glycolytic

enzymes and, consequently, low rates of glucose utilization (Hertz and Dienel, 2002).

Glucose oxidative metabolism is the main source of energy to the brain, being tightly

regulated to generate ATP and provide carbon atoms for biosynthetic reactions in

conjunction with local functional brain activities. Pyruvate oxidation through the TCA cycle

originates a high yield of ATP via the ECT and establishes a link between bioenergetics

and amino acid pools. At steady state, more than 90% of the available glucose undergoes

oxidative degradation in the whole brain. In fact, brain activation is associated to

increased glucose utilization (that can sometimes be higher than the concomitant increase

in oxygen consumption. The exact mechanisms and type of cells involved in this

“metabolic mismatch” are still not fully understood, but some authors have suggested that

it may include the accumulation and/or the release of lactate and eventually the

generation of other incompletely oxidized glucose metabolites (Hertz and Dienel, 2002).

Regions functionally more active and consequently, with increased metabolic rate of

glucose, exhibit greater capacity for fuel delivery and recruitment of the related

components (as transporters and enzymes) in order to match the local metabolic needs

with adequate fuel delivery (Hertz and Dienel, 2002). In the last years, in vivo imaging

studies such as positron emission tomography (PET) using an analog of the glucose

molecule labeled with a radioactive isotope (2-deoxy-2-[18F]fluoro-D-glucose, 18F-FDG)

have provided valuable information concerning glucose metabolism in the brain in resting

as well as activated conditions and cellular participation in the bioenergetics of brain

activation.

Besides being a source of energy, glucose oxidation also results in the production of

important neurotransmitters, such as glutamate, an amino acid neurotransmitter that is

produced by the transamination of α-ketoglutarate (an intermediate of the TCA cycle).

Another important neurotransmitter, derived from glutamate (and therefore from glucose)

is γ-aminobutyric acid (GABA). The first step of the glucose oxidation occurs in the cytosol

(glycolysis) and the subsequent steps take place into the mitochondria (PDHC, TCA cycle

and ETC). The TCA cycle is important for amino acids metabolism as well as for the

conversion of sugar metabolites into fragments that are then converted into amino acids

by the transamination. In some conditions, brain can also obtain energy from its modest

glycogen storage (located mainly in astrocytes), which may supply the needed fuel when

metabolic demands exceed the supply of glucose from the blood (Blass, 2002).

Page 48: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

48

The passage of glucose from the blood to the brain cells requires the action of several

isoforms of the glucose transporters. Endothelial cells forming the blood brain barrier

(BBB) express the glucose transporter GLUT1 and neurons express the isoform GLUT3.

There is no evidence that GLUT3 is expressed in other brain cells than neurons

(astrocytes, oligodendrocytes or endothelial cells) (Simpson et al., 2008). GLUT5 is

expressed in the microglia (Maher et al., 1994; Vannucci et al., 1997). Although glucose is

the main energy source, brain cells are able to use other sources to produce energy, as

fatty acid oxidation by the astrocytes. Additionally, ketone bodies can be used as a fuel by

neurons, astrocytes and oligodendrocytes (Edmond, 1992). Whereas stimulation of brain

activity increases glucose utilization in specific brain areas, activity inhibition or neuronal

loss reduces it. Glycolysis and glycogenolysis are stimulated in astrocytes by glutamate,

released in the course of neuronal activity (Magistretti et al., 1994; Pellerin and Magistretti,

1994). Glucose is metabolized to lactate and together with glucose itself, is released into

the extracellular space and taken up by neurons, being used as a substrate for oxidative

phosphorylation (Dringen et al., 1993). If glucose becomes scarce, astrocytes are able to

supply ketone bodies, because in these conditions, glucose is too precious to serve as a

fuel (Randle et al., 1978). However, the brain is the last organ to use substantial ketone

bodies metabolism after long periods of starvation (Heininger, 2002a). Although neurons

cannot use fatty acids, astroglial cells can degrade them by β-oxidation, providing ketone

bodies to the neurons (Edmond, 1992). By one side, oxidative metabolism yields much

higher amounts of ATP, but by the other side, it represents an increased risk of oxidative

stress, contributing to the enhanced vulnerability of neurons (Almeida et al., 2001).

It has been shown that the levels of activity of the enzymes involved in glucose

metabolism not only vary significantly, but also do not distribute uniformly in the brain,

indicating that glucose metabolism may present cellular compartmentalization in the brain,

with high rates of glycolysis in astroglia and high rates of PDHC, TCA cycle and electron

transport in neurons (Blass, 2002). Moreover, it has been reported that the activity of key

enzymes for energy metabolism is reduced in neurodegenerative diseases. They include

the PDHC, the α-ketoglutarate dehydrogenase complex (KGDHC) and the cytochrome

oxidase (COX) (Blass, 1999; Gibson et al., 2005). Abnormalities in mtDNA can occur, at

least in part, as a result of oxidative stress. Additionally, ECT complexes III, IV and V are

also impaired in neurodegenerative diseases (table 1.4).

Heininger defined aging as a genetically programmed deprivation syndrome in which the

soma, stressed by the degeneration of the trophic milieu, develops mechanisms to find or

maintain survival pathways, characterized by hypometabolism, adjustments of the

Page 49: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

49

glucose-fatty acids cycle, oxidative stress and modulation of DNA repair (Heininger,

2002b). The adaptive response of the glucose-fatty acids cycle occurs during the normal

process of brain aging and results in the reduction of both cerebral blood flow and glucose

utilization (Heininger, 1999), the mismatch of oxygen and glucose consumption, the

increase use of ketone bodies as a fuel (Heininger, 2002a) and the decrease of brain

insulin receptor densities and signal transduction (Frolich et al., 1999). Moreover, during

aging, degeneration of proglycolytic neurotransmitters (acetylcholine, noradrenaline and

serotonin) is observed (Heininger, 1999).

The brain is highly susceptible to oxidative damage because it presents a high oxygen

consumption rate, it is rich in easily peroxidizable unsaturated fatty acids and relatively

low content in antioxidant enzymes (Nunomura et al., 2006; Gandhi and Abramov, 2012).

Moreover, three main reasons contribute for mtDNA increased susceptibility to oxidative

damage: a) it is not protected by histones, making their mutation rate ten times higher

than nuclear DNA (Filosto et al., 2011), b) it is too close to the source of ROS, the ETC, c)

less efficient repair machinery (Petrozzi et al., 2007). On the other hand, calcium

homeostasis deregulation can lead to intracellular calcium overload, disrupting proton-

motive force and consequently, inducing mitochondrial permeability transition (MPT),

which, once activated, results in the release of cytochrome c and caspase-mediated

apoptosis (Lin and Beal, 2006).

Oxidative stress has been considered one of the most important factors associated to

neurodegeneration, in which there is an imbalance between the production of ROS or

reactive nitrogen species (RNS) and its inactivation by cellular antioxidant machinery,

resulting in cell damage. ROS and RNS attack key molecules, causing DNA, RNA and

protein oxidation and lipid peroxidation (Gandhi and Abramov, 2012). More recent data

suggest that ROS and RNS are involved in signaling cascades, regulating normal cell

function (Droge, 2002; Valko et al., 2007). It has been shown that many of the ROS-

mediated responses can protect the cells against oxidative stress and reestablish the so-

called redox homeostasis (Droge, 2002; Valko et al., 2007). The term ROS encloses both

(free) radicals (substances that have unpaired electrons) and chemicals that participate in

radical type reactions (substances that gain or lose electrons, but are not considered true

radicals as they do not have unpaired electrons) (Magder, 2006). Examples of free

radicals are atomic hydrogen (with only one electron), molecular oxygen (O2, with two

unpaired electrons in the outer orbitals). Partially reduced forms of oxygen are highly

unstable because they must react either by accepting or donating electrons, originating

Page 50: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

50

free radicals as superoxide (O2�-) and hydroxyl radical (OH�) (Magder, 2006; Gandhi and

Abramov, 2012).

Examples of non-radical ROS are hydrogen peroxide (H2O2), hypochlorous acid (HOCl),

ozone (O3) and singlet oxygen (1O2). In addition to the oxygen radicals, there are also

nitrogen-based radicals such as nitric oxide (NO), nitric dioxide (NO2), peroxynitrite

(ONOO-), sulfur-based radicals such as thinyl (RS) and perthinyl (RSS), carbon-based

radicals as trichloromethyl (CCl3�) (Magder, 2006).

Due to their instability, ROS or RNS establish chemical reactions with biological

molecules, inducing changes in cell functions that can, ultimately, cause cell death.

Because of its unique features, brain cells and neurons in particular, need a robust and

efficient antioxidant system. In fact, their increased vulnerability to oxidative stress is

essentially due to three main factors (Gandhi and Abramov, 2012): a) greater oxygen

consumption when compared to other tissues (around 10-fold); b) neurons are non-

dividing, and long living cells, presenting cumulative exposure to all kind of insults; c)

presence of nitric oxide, having physiological role in the brain, which can originate RNS

such as ONOO-, in the presence of O2�- (Gandhi and Abramov, 2012).

ROS produce deleterious effects in living organisms through its actions at cellular and

molecular level. The origin of these species can be exogenous (produced from

xenobiotics, chemical toxins or radiation exposure) or endogenously produced (from ETC,

cytochrome P-450) (Cadet and Brannock, 1998). They attack lipid molecules from cell

membranes, causing lipid peroxidation, changing membrane fluidity and shape. This

damage disrupts calcium homeostasis, fundamental for intracellular signaling cascades.

Moreover, peroxides can also cause damage to other key molecules such as proteins and

DNA (Cadet and Brannock, 1998; Magder, 2006). Oxidative stress conditions favor

Fenton reaction, in which iron reduction by O2�- originates the highly toxic radical OH�

(Thomas et al., 2009). Activation of Poly(ADP) polymerase (PARP) is the result of the

indirect damage resulting from ROS, leading to changes in gene expression, ATP

depletion and, ultimately, to apoptosis (Zingarelli et al., 1996). Other important targets of

ROS are proteins that undergo oxidation, disturbing receptors, enzymes, cell signaling

pathways and the production of carbonyl groups.

Under normal physiological conditions, electron leakage from the ETC is the major source

of O2�-. About 1-3% of mitochondrial reduced O2 is in the form of O2

�- originated at

complexes I and III. Additionally, some enzymes are also able to produce O2�- in the

course of their normal functions, such as amino acid oxidase, cytochrome oxidases,

Page 51: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

51

monoamine oxidases, xanthine oxidase and aldehyde oxidase (Cadet and Brannock,

1998; Magder, 2006). Other known sources of O2�- are: the autoxidation of

catecholamines, reperfusion after ischemia episode and the stimulation of glutamate

receptors (Cadet and Brannock, 1998). In the brain, catecholamines are indeed an

important source of free radicals as its autoxidation induces the production of quinones

and, subsequently, the generation of H2O2 and O2�- radicals and damage to proteins.

Moreover, the breakdown of dopamine, serotonin and norepinephrine catalyzed by

monoamine oxidase, originates H2O2, as shown in figure 1.6.

Figure 1.6 – Free radical production from catechola mines breakdown and autoxidation and

glutamate receptors . Legend: Ca++ - calcium ion; DA - dopamine; Fe++ - iron ion; Glu - glutamate;

GSH-Px - glutathione peroxidase; H2O2 - oxygen peroxide; L-Arg - L-arginine; MAO - monoamine

oxidase; NO - nitric oxide; O2�- - superoxide radical; OHDA - 6-hydroxydopamine; ONOO -

peroxynitrite; SOD - superoxide dismutase; [From (Cadet and Brannock, 1998), with permission]

Although the idea that mitochondria are the major source of ROS is generally accepted, in

fact, under pathological circumstances cytosolic enzymes as nicotinamide adenine

dinucleotide phosphate (NADPH)-oxidase produce much higher amounts of free radicals

than mitochondria (Gandhi and Abramov, 2012). Furthermore, more recently, Brown and

Borutaite have demonstrated that other organelles such as the endoplasmic reticulum and

peroxisomes have a greater capacity to produce ROS than mitochondria (Brown and

Borutaite, 2011). Because they are continuously exposed to free radicals, mitochondria

have a very good antioxidant system. Overproduction of ROS by one side and suboptimal

Page 52: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

52

response by the antioxidant systems by the other, induces oxidative stress and

neurodegeneration (Gandhi and Abramov, 2012). Neurodegenerative diseases are

characterized by a progressive and selective loss of neurons, anatomically or

physiologically correlated (Lin and Beal, 2006). Aging is the major risk factor for the onset

of the most common neurodegenerative diseases (such as Alzheimer’s, Parkinson’s and

Huntington’s diseases) in which mitochondria have been implicated (Lin and Beal, 2006;

Gandhi and Abramov, 2012). A very brief discussion about the involvement of

mitochondria in these pathologies will be presented (table 1.4).

Since we live in oxygen rich environments and, as mentioned above, ROS are generated

in the course of normal metabolic functions, cells were forced to develop defense

mechanisms in order to guarantee homeostasis and health status. The existence of

effective scavenging systems makes that oxygen radicals have very short lives (Cadet

and Brannock, 1998; Valko et al., 2007). There are different types of defense mechanisms

against oxidative stress that may act at different phases concerning oxidative injury,

namely preventive and repair mechanisms, physical barriers and antioxidant defenses

(Valko et al., 2007). The enzymes involved in antioxidant defenses are: superoxide

dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT). Examples of

non-enzymatic antioxidants are: vitamins C and E, glutathione (GSH), carotenoids and

flavonoids (Magder, 2006; Valko et al., 2007).

GSH is a tripeptide thiol particularly abundant in the cytosol, nuclei and mitochondria,

exerting its antioxidant action by working as a cofactor of several detoxifying enzymes, by

scavenging OH� and 1O2, by regenerating other antioxidants to their active form and by

working in conjunction with other antioxidants as GSH-Px (Magder, 2006; Gandhi and

Abramov, 2012). SOD exerts its action particularly against O2�-, existing in three different

forms: the cytosolic and releasable copper-zinc SOD (CuZn-SOD) and the mitochondrial

manganese SOD (MnSOD). CAT exists in the peroxisomes, lysosomes and mitochondria,

neutralizing hydrogen peroxide to water and molecular oxygen, in conjunction with GSH-

Px. The latter also constitutes another line of defense against organic hydroperoxides and

prevents lipid peroxidation, membrane destabilization and membrane dysfunction (Cadet

and Brannock, 1998). Vitamins E and C and β-carotene are absorbed from diet, mostly

from fruits and vegetables, constituting important lines of defense in the brain. Vitamin E

traps peroxyl radicals, originating tocopheroxyl radicals which are then reduced back to

the original tocopherol by vitamin C. Moreover, vitamin C reacts with ROS, originating

dehydroscorbate, which is reduced by GSH (Cadet and Brannock, 1998).

Page 53: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

53

In fact, ROS and RNS are physiological products from normal cellular metabolism, able to

induce cell damage or work as secondary messengers in several cell signaling cascades.

Ironically, these reactive species may also participate in a number of redox-regulatory

mechanisms and protect cells against oxidative stress and maintenance of the so called

redox homeostasis. However, an imbalance between ROS or RNS production and its

inactivation by the cellular antioxidant machinery, results in cell damage and ultimately in

cell dysfunction and death (Cadet and Brannock, 1998; Valko et al., 2007).

Carnitines and ALC in particular have been reported to have antioxidant properties,

namely by increasing the endogenous pool of GSH (Aureli et al., 1999) and promoting the

activity of antioxidant enzymes (El-Awady el et al., 2010; Hao et al., 2011; Haorah et al.,

2011). Lower doses of ALC seem to be more efficient in preventing oxidative stress (Liu et

al., 2002). As an example, ALC improved age-related decline in mitochondrial cristae in

the hippocampus, while higher doses not only did not decreased lipid peroxidation in the

brain, but also increased protein carbonyl content. Based on these results, the authors

suggest that increased oxidative stress may be a side effect of high doses of ALC (Liu et

al., 2002).

Page 54: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

54

Table 1.4 – Main neurodegenerative diseases and mit ochondria involvement

Disease Characterization Clinical

Symptoms Evidence of Mitochondrial Involvement References

Alzheimer’s

Disease

- Late onset and progressive

disease, characterized by neuronal

loss, particularly in the cortex and

hippocampus.

- Presence of extracellular senile

plaques (containing the peptide β-

amyloid) and neurofibrillary tangles

(containing hyperphosphorylated of

the microtubular protein tau).

Cognitive

functions

deterioration with

behavioral and

personality

alterations.

- Disruption of several mitochondrial enzymes such as PDHC, KGHC,

COX, results in decreased brain metabolism and function.

- Aβ accumulates in damaged mitochondria, impairing ETC and inhibiting

mitochondrial enzymes (e.g. COX, KGDHC). These events lead to MPT

opening and trigger the apoptotic cascade.

- Complex I and COX inhibition promotes tau phosphorylation and thus

facilitates microtubules aggregation. This leads to the formation of

tangles, which consist in microtubules aggregates with

hyperphosphorilated tau protein.

- mtDNA mutations, alterations in mitochondrial fission and fusion

processes as well as morphological changes in mitochondria have also

been reported.

(Pereira et al.,

1998; Blass,

2002; Coskun

et al., 2004;

Virmani and

Binienda, 2004;

Lin and Beal,

2006; Petrozzi

et al., 2007;

Valko et al.,

2007; Wang et

al., 2008;

Filosto et al.,

2011)

Parkinson’s

Disease

- Late onset and progressive

disease, characterized by loss of

dopaminergic neurons in SNPC

and the accumulation of inclusions

positive for α-synuclein and

ubiquitin (Lewy bodies).

Rigidity,

bradykinesia and

tremor

- Accumulation of point mutations and deletions in mtDNA in PD brains.

- Inhibition of mitochondrial respiratory complex I in the SNPC.

- Six out of nine nuclear genes that have been associated to PD are

directly or indirectly linked to mitochondrial function, marked in blue: α-

synuclein, DJ-1, leucine-rich-repeat kinase 2 (LRRK2), parkin,

phosphatase and tensin homologue (PTEN)-induced kinase 1 (PINK1),

nuclear receptor NURR1, HTRA2, tau and ubiquitin carboxy-terminal

hydrolase L1.

- α-synuclein overexpression impairs mitochondrial function by reducing

(Blass, 2002;

Lin and Beal,

2006; Petrozzi

et al., 2007;

Valko et al.,

2007; Chinta et

al., 2010;

Filosto et al.,

2011)

Page 55: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

55

complex I activity and increasing free radical production.

- DJ-1 translocates to the mitochondrial matrix, downregulating PTEN-

tumour suppressor, protecting cells from oxidative stress.

- Parkin located at OMM inhibits cytochrome c release; it may also

interact with TFAM, regulating mitochondrial biogenesis and mitophagy.

- PINK1, a mitochondrial kinase protects against apoptosis, suppressing

cytochrome c release. PINK1 deficiency leads to mitochondrial calcium

overload and, in combination with ROS, initiates opening of MTP.

- Mutations in the above mentioned proteins have been associated to PD.

Huntington’s

Disease

- Genetic autosomic disease

characterized by loss of

GABAergic neurons in the striatum,

resulting in the progressive atrophy

of the caudate nucleus, putamen

and globus pallidus.

- Caused by the expansion of CAG

trinucleotide repeat in the

huntingtin (HTT) gene, expanding

the polyglutamine stretch in the

corresponding protein, with

subsequent induced toxicity.

Progressive

choreiform

movements,

psychiatric

disturbances and

dementia.

- Reduced activity of ETC complexes II, III and IV.

- High levels of lactate in the cortex and basal ganglia.

- Huntingtin associates with OMM, increasing sensitivity to calcium-

induced mitochondrial permeabilization and cytochrome c release.

- Mutant huntingtin binds to p53 (tumour suppressor that regulates genes

involved in mitochondrial function and oxidative stress), translocating it to

mitochondria, causing subsequently Bax activation.

(Blass, 2002;

Bae et al.,

2005; Lin and

Beal, 2006;

Petrozzi et al.,

2007; Filosto et

al., 2011)

Legend: Aβ – β-amyloid; COX – cytochrome-c oxidase; ETC – electron transport chain; KGHC- ketoglutarate dehydrogenase complex; mtDNA – mitochondrial DNA; MPT –

mitochondrial pore transition; OMM – outer mitochondrial membrane; PD – Parkinson’s disease; PDHC – pyruvate dehydrogenase complex; SNPC – substantia nigra pars

compacta; TFAM – mitochondrial-transcription factor A.

Page 56: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

56

1.8. METHAMPHETAMINE AS AN OXIDATIVE STRESS INDUCER

Methamphetamine (N-methyl-O-phenylisopropylamine, METH) is a psychostimulant and a

highly addictive drug, abused worldwide, particularly popular among the youngest due to

its acute effects, namely the onset of euphoria, the improvement of alertness and

concentration, the sensation of well-being and self-confidence, the increased libido and

decreased appetite (Barr et al., 2006; Fleckenstein et al., 2007). However, a long list of

acute side effects causing serious health problems may also occur, partially due to the

effects of METH on epinephrine and norepinephrine release by the adrenal glands:

tachycardia, increased blood pressure, hyperthermia, muscle rigidity and ataxia, organ

failure (liver and kidneys), nausea, blurred vision, anxiety, psychosis and seizures (Barr et

al., 2006; Cadet et al., 2007). Human and animal studies have shown that withdrawing the

administration of high doses of amphetamines originates the opposite effects seen on

acute administration, leading to the onset of fatigue, anxiety, anhedonia, depression and

lack of concentration (Barr et al., 2006). Chronic administration of amphetamines is

associated with attention and memory deficits, impaired learning capacity, as well as

compromised decision making (Cadet et al., 2007). Moreover, schizophrenia has been

associated with METH abuse (McKetin et al., 2010).

Under physiologic conditions neuronal activation induces vesicular dopamine release in

the synaptic cleft, that will be then removed by dopamine transporters (DAT) and stored

again in the vesicles by the vesicular monoamine transporter 2 (VMAT-2), thus protecting

DA from oxidation. However, in the presence of METH, DA homeostasis will be affected in

a variety of ways which will be described below.

Due to its relatively high lipophilicity, METH crosses the blood brain barrier (BBB) very

easily (Barr et al., 2006; Fleckenstein et al., 2007), and after entering monoaminergic

terminals by DA or serotonin transporters, it causes the release of monoaminergic

neurotransmitters as dopamine, norepinephrine and serotonin, which will be oxidized to

produce ROS, resulting in neuronal damage and death (Kuczenski et al., 1995; Davidson

et al., 2001; Sulzer et al., 2005). METH belongs to the group of amphetamine-type

stimulants, which are synthetic compounds including amphetamine (1-methyl-2-

phenylethylamine), METH itself and MDMA (Fleckenstein et al., 2007). All these

compounds share similarities in their chemical structure with the monoaminergic

neurotransmitter dopamine (DA), as shown in figure 1.7 (Fleckenstein et al., 2007). This is

the reason why METH is able to compete so efficiently with DA for access to DAT. The

latter, expressed in the dopaminergic neurons, are responsible for DA reuptake after each

Page 57: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

57

round of neurotransmission, in order to store it again into new DA vesicles for future

events of excitation.

Figure 1.7 – Molecular structure of the most common amphetamines and its similarities with

dopamine . [From (Fleckenstein et al., 2007)]

Concerning the effects on catecholamine-containing neurons, psychostimulants can be

categorized into two classes: amphetamines, that inhibit the reuptake of catecholamines

and are potent releasers and non-amphetamines (e.g. cocaine), that inhibit reuptake but

are more limited in their releasing capacities (Ritz et al., 1987). Amphetamines cause DA

release from the synapses because they are weak bases (the “weak base hypothesis”),

highly permeable, able to enter into vesicles and bind to free protons. This induces

alkalization of the existing acidic pH gradient and decreases the energy that provides

accumulation of neurotransmitters in the cytosol and in the synaptic cleft (Sulzer et al.,

2005). Additionally to this redistribution of catecholamines from the vesicles to the cytosol,

it was demonstrated that amphetamines not only reverse the activity of monoamine

transporters, but also decrease the expression of DAT at the cell surface (Saunders et al.,

2000; Barr et al., 2006). Other mechanisms involved in the increase of the cytosolic levels

of monoamines include the inhibition of the monoamine oxidase (MAO) activity and the

increase in the expression and activity of tyrosine hydroxylase (TH), the DA-synthesizing

enzyme (Mandell and Morgan, 1970; Mantle et al., 1976; Sulzer et al., 2005). Altogether,

these mechanisms (represented in figure 1.8) contribute to the powerful action of

amphetamines on monoamines release, presenting prolonged behavioral effects

comparatively to other psychostimulants due to its longer biological half-life (8-13h for

METH and 1-3h for cocaine) (Barr et al., 2006).

Page 58: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

58

Figure 1.8 – Mechanisms through which METH increase s synaptic levels of catecholamines .

The represented mechanisms involve: (1) the redistribution of catecholamines from the vesicles

into the cytosol and the reverse transport through plasma membrane transporters; (2) the

impairment of monoamine transporters activity; (3) the decrease in the expression of DA

transporters at the cell surface; (4) the inhibition of MAO activity; (5) the increase in the expression

and activity of tyrosine. The cytosolic dopamine is then oxidized originating reactive oxygen

species. DA – Dopamine; DAT – Dopamine transporter; MAO – Monoamine oxidase; ROS –

Reactive oxygen species; VMAT - vesicular monoamine transporter. [Adapted from (Barr et al.,

2006)]

Abnormalities in the dopaminergic system are a common characteristic of METH-induced

toxicity as well as Parkinson’s disease (Brotchie and Fitzer-Attas, 2009; Yagi et al., 2010)

and schizophrenia (Akiyama et al., 1994; Kurachi, 2003). The nigrostriatal DA pathway is

the most affected by METH-induced neurotoxicity comparing to the mesocorticolimbic

dopaminergic projections from the ventral tegmental area to the forebrain regions, such as

the nucleus accumbens (Eisch et al., 1992; Cass, 1997; Barr et al., 2006). Multiple high-

doses of METH administration cause a greater impact on the DA depletion or on the

reduction in DAT density in the striatum rather than in the nucleus accumbens, making the

former more sensitive than the nucleus accumbens to neurotoxic effects of METH (Eisch

et al., 1992; Cass, 1997). Additionally, a region-specific study has shown that the dorsal

caudate putamen is less vulnerable to dopaminergic deficits than the ventral caudate

putamen region (Eisch et al., 1992; Cass, 1997). There is extensive evidence about the

involvement of DAT in METH neurotoxicity as the administration of DAT inhibitors protects

DD

ROS

Page 59: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

59

against METH toxicity (Marek et al., 1990b, a; Pu et al., 1994). Moreover, heterozygotic

DAT knock-out mice show resistance to dopaminergic deficits induced by METH (Cubells

et al., 1994; Fumagalli et al., 1998). Under normal physiologic conditions, DAT are

responsible for the transport of extracellular DA back into the nerve terminals. After METH

administration, however, a reduction in DAT activity and/or cell surface localization may

occur, decreasing DA efflux, promoting its oxidation (Riddle et al., 2006). It has been

reported that a single high dose of METH reversibly decreases DAT function, as a result

of phosphorylation and internalization of the transporter (Saunders et al., 2000). Although

the exact mechanisms involved in the increased vulnerability of the striatum are not

known, some experiments have shown that the striatum has a significantly higher density

and activity of DAT (Eisch et al., 1992; Povlock and Schenk, 1997).

Other experiments based on the administration of TH inhibitors (α-methyl-p-tyrosine)

(Gibb and Kogan, 1979; Uehara et al., 2004; Fleckenstein et al., 2007; Thomas et al.,

2008) or DA receptor antagonists (chlorpromazine or haloperidol) (Sano et al., 1982;

Nagai et al., 2007; Oshibuchi et al., 2009) shown to prevent METH-induced deficits,

indicating that DA itself is also involved in METH-induced toxicity. As mentioned before,

METH induces an abnormal accumulation of DA in the cytosol, which, in the presence of

METH is rapidly oxidized into ROS (figure 1.8) as quinones and semiquinones, in such

amount that exceeds the capacity of the antioxidant systems, thus damaging DA-

containing neurons (De Vito and Wagner, 1989; Cubells et al., 1994; Cadet and Brannock,

1998; Sulzer and Zecca, 2000). Striatal extracellular levels of DA are four to five fold of

those in nucleus accumbens, reinforcing the increased vulnerability of the striatum

(Hernandez et al., 1987; Cass, 1997). Furthermore, accumbal antioxidant levels

(particularly superoxide dismutase) are more than twice those of striatum, contributing to

the differential vulnerability to METH of these brain regions (Perumal et al., 1992;

Kunikowska and Jenner, 2002). The VMAT-2 is the responsible for the sequestration of

cytosolic DA into the vesicles until posterior release and by doing its function is also

contributing for the protection of dopaminergic neurons from DA oxidation and thus, from

oxidative damage (Riddle et al., 2006; Fleckenstein et al., 2007; Sulzer, 2011). However,

the presence of METH seems to induce a subcellular redistribution of VMAT-2 such that

fewer transporter associated to storage vesicles will be available to capture DA,

preventing its subsequent oxidation (Riddle et al., 2006).

It is reported that RNS are formed concomitantly to ROS as a result of DA oxidation. The

major cellular consequences from these oxidative species are lipid peroxidation and the

production of protein carbonyls (Yamamoto and Zhu, 1998; Wu et al., 2007; Alves et al.,

Page 60: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

60

2009). In vivo studies show that a single administration of METH to Wistar rats, induced

mitochondrial oxidative damage both in the striatum and cortex (Shiba et al., 2011).

Repeated doses of METH (4 x 10mg/kg/injection 2h apart) resulted in increased oxidative

stress markers in the striatum and hippocampus in mice (Gluck et al., 2001). Despite this

observation, the cellular antioxidant systems exhibited increased activity as the same

dose scheduling induced an early (2h) increased in GSH and oxidized GSH in Sprague–

Dawley rat striatum (Harold et al., 2000). Furthermore, RNS play a major role in METH-

induced dopaminergic neurotoxicity. Both in vitro experiments using PC12 cells and in

vivo studies in mice, demonstrated a significant increase in the production of 3-

nitrotyrosine (3-NT) induced by METH, paralleled by DA depletion, respectively in PC12

cells and in mice striatum (Imam et al., 2001b).

Human postmortem studies of METH abusers displayed increased activity of antioxidants:

CuZn-SOD and oxidized GSH levels increased 14% and 58%, respectively. Additionally,

in METH abusers with severe DA loss, a tendency for decreased GSH levels was

observed. Authors suggest that antioxidant systems may be preserved in METH

administration and that GSH depletion occurs only as a result of severe dopamine loss

(Mirecki et al., 2004).

In vitro studies using dopaminergic cell lines shown that METH induces a temporal

sequence of several cellular events (figure 1.9), from decreased mitochondrial membrane

potential to apoptosis (Wu et al., 2007).

Figure 1.9 – Temporal sequence of events associated to METH in vitro toxicity .

Legend: � - decreased; �� - strongly decreased; � - increased; ROS – reactive oxygen species;

mtDNA - mitochondrial deoxyribonucleic acid

1h

� Mitochondrial membrane potential

8h

�� Mitochondrial membrane potential

� ROS

24h

� Mitochondrial mass

� mtDNA copy number

48h

Apoptosis METH

Page 61: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

61

As shown in figure 1.9, mitochondria are heavily involved in METH toxicity, being a known

source of ROS. Antioxidants and free radical scavengers have beneficial effects in the

reduction of METH-induced oxidative damage (Perumal et al., 1992; Dhitavat et al., 2005;

Alves et al., 2009; Koo et al., 2011). METH causes impairment of mitochondrial function

by inhibiting the complexes I (Virmani et al., 2004), II, III (Brown et al., 2005) and IV

(Burrows et al., 2000) of the ETC and this is likely to increase the production of free

radicals, resulting in oxidative damage (Brown and Yamamoto, 2003) and energy

depletion due to decreased ATP generation. Other mechanisms for METH-induced

neurotoxicity may comprise the activation of apoptotic cascades, involving caspases, pro-

apoptotic genes from Bcl-2 family and the tumor suppressor gene p53 (Imam et al.,

2001a; Koo et al., 2011). A single high dose of METH (40 mg/kg) induced apoptosis in

monoaminergic cells from the frontal cortex by overexpressing pro-apoptotic proteins

(Bax, Bad and Bid) and decreasing the expression of anti-apoptotic Bcl-2-related proteins

(Bcl-2 and Bcl-XL) (Jayanthi et al., 2004). At mitochondrial level, a cascade of events

occurs, namely the early release (30 minutes post METH injection) of apoptosis-inducing

factor (AIF), smac/DIABLO (a pro-apoptotic molecule working as an indirect activator of

caspases through the inhibition of the inhibitor of apoptosis proteins, IAPs) and

cytochrome c, with subsequent activation of caspases (Du et al., 2000; Imam et al., 2003;

Jayanthi et al., 2004). Other proteases such as calpain (a calcium-dependent enzyme) are

also involved in METH-mediated neuronal cell degeneration, as shown in human

neuroblastoma SH-SY5Y cultured cells (Suwanjang et al., 2010). Calpain exists as an

inactive pro-enzyme in the cytosol, being activated in response to excessive calcium influx

and thus able to cleave several proteins from the cytoskeleton, other proteases and

signaling molecules (Suwanjang et al., 2012).

More recently, the involvement of both pro- and anti-inflammatory immune mediators has

been demonstrated subsequently to METH administration (Itzhak and Achat-Mendes,

2004; Gonçalves et al., 2008; Tocharus et al., 2010; Pendyala et al., 2012).

The role of environmental and body temperature in METH toxicity is still a topic of

discussion due to controversial findings. Ali et al. reported that multiple doses of METH (4

x 10 or 20 mg/kg) administered in a cold room (4ºC) decreased significantly striatal

dopamine and serotonin depletion, observed when the same doses were given in a 23ºC

room (Ali et al., 1994; Baucum et al., 2004). The underlying mechanism might somehow

interact with glutamate neurotransmission. The administration of a glutamate receptor

antagonist (NMDA), which prevents body temperature to rise, induced a protective

mechanism over striatal DA and serotonin depletion (Bowyer, 1995). Conversely, more

Page 62: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

62

recently, studies have shown that mechanisms underlying METH neurotoxicity are

temperature-independent (Thomas et al., 2008).

Although research has been focused primarily on the neurotoxicity of METH in

dopaminergic neurons, lately there has been an increasing interest on the effects of

METH upon serotoninergic terminals. Similar findings were obtained, in which high doses

of METH caused significant loss of serotoninergic terminals, with changes in the levels of

serotonin and serotonin transporters (Gross et al., 2011; Weng et al., 2012). However,

effects on serotonin neurons are more diffuse, including regions as the hippocampus and

perirhinal cortex, anterior cingulate cortex, caudate nucleus, nucleus accumbens and

septum (Armstrong and Noguchi, 2004; Kish et al., 2009).

Increased levels of glutamate (the main excitatory neurotransmitter in mammals) in

several brain areas as the striatum, the hippocampus and the ventral tegmental area

(VTA) subsequently to METH administration, as well as changes in glutamate

transporters, suggest the involvement of the neurotransmitter in METH toxicity (Mark et

al., 2007; Kerdsan et al., 2012). Neuronal excitotoxic damage occurs due to excessive

glutamate release and activation of glutamate receptors (Cadet et al., 2003). Moreover, a

single dose of 30 mg/kg induced significant changes in the striatal

glutamatergic/GABAergic homeostasis, paralleled by impaired mice motor activity (Pereira

et al., 2012). Striatal dopaminergic cells present the receptors α-amino-3-hydroxy-5-

methyl-4-isoxazole propionic acid (AMPA) and N-methyl-D-aspartate (NMDA), which are

activated by glutamate, promoting Ca2+ influx into the neuron, leading to calcium overload

that is damaging to the mitochondria, and ultimately to the cell (Schinder et al., 1996).

Finally, more recently, it has been demonstrated that the BBB plays a key role in METH-

induced neurotoxicity. METH acts either by inducing a severe disruption of BBB integrity

in several brain regions (as the cortex, thalamus, hypothalamus, hippocampus, amygdala

and cerebellum) or by increasing BBB permeability (Kiyatkin and Sharma, 2009; Ramirez

et al., 2009). Possible mechanisms involve decreasing the levels of tight-junction proteins

in the hippocampus and increasing activity and expression of matrix metalloproteinases

(Martins et al., 2011), increasing glial activation (Snider et al., 2012), increasing enzyme

activation related to BBB cytoskeleton remodeling (Kousik et al., 2012) and the induction

of neuroinflammatory pathways (Gonçalves et al., 2008; Kousik et al., 2012).

Animal experiments have elucidated that METH effects are largely dependent on dose

schedule (the number of doses, the amount per dose and the time interval between

doses). Extrapolating the results obtained in animals to humans may reveal a difficult task

Page 63: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

63

due to the fact that animal studies are mainly based on acute exposure to multiple high

doses of METH during one or two days (which may cause 30-60% loss of dopaminergic

terminals), but human studies are based on months or years of exposure (Barr et al.,

2006). This question was well addressed by Krasnova et al. (Krasnova et al., 2010).

Additionally, the degree of purity of administered drugs might be rather different. Human

neuroimaging studies have been invaluable in the characterization of cerebral alterations

induced by METH abuse both at functional (positron emission tomography, PET and

functional magnetic resonance imaging, fMRI) and morphological level (magnetic

resonance imaging, MRI). Major morphological imaging findings include loss of gray

matter in the cingulate, limbic and paralimbic cortices, hippocampus atrophy and white

matter hypertrophy (Thompson et al., 2004; Daumann et al., 2011; Nakama et al., 2011;

Orikabe et al., 2011). Concerning function, PET studies revealed significant decreases in

dopamine-relevant proteins in the orbitofrontal cortex, dorsolateral prefrontal cortex,

striatum, nucleus accumbens and amygdala, which presented a good correlation with

cognitive impairments and the severity of psychiatric symptoms (Volkow et al., 2001b;

Volkow et al., 2001c). The density of dopamine D2 receptors in the striatum is significantly

reduced in METH abusers (Volkow et al., 2001d). More recently this loss of dopamine D2

receptors (figure 1.10) has been associated with decreased glucose metabolism by the

brain (figure 1.11) (Gouzoulis-Mayfrank et al., 1999), which over time, tends to substitute

glucose as the main source of energy by acetate in some brain areas. These findings

about changes on brain metabolism are also applicable to other addictive substances

such as alcohol and cocaine (Volkow et al., 2006; Volkow et al., 2013).

Figure 1.10 – Positron emission tomography images o f dopamine D2 receptors . Images show

the uptake of 11C-raclopride by dopamine D2 receptors in the striatum of a healthy volunteer and a

METH abuser. Note the significant loss of D2 receptors in the METH abuser. [From (Volkow et al.,

2001d)]

Page 64: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

64

Figure 1.11 – 18F-FDG PET images depicting brain glucose uptake und er control condition or

after METH administration. Axial 18F-FDG-PET images of subjects under control or METH

administration while performing a word repetition task. Note the decreased cortical and increased

cerebellar activity in the METH subject. [From (Gouzoulis-Mayfrank et al., 1999), with permission]

The rewarding effects of METH and drugs of abuse in general are due to their ability in

increasing DA, particularly in the nucleus accumbens. Thus, the mesolimbic DA pathway,

which involves cells in the VTA that projects to the nucleus accumbens, plays an

important role in drug reward. The mesostriatal (DA cells in substantia nigra that project to

the dorsal striatum) and mesocortical (DA cells in VTA that project to the frontal cortex)

are also involved in mechanisms of drug reward and addiction (Wise, 2009; Volkow et al.,

2011). METH is able to increase DA-evoked long-term adaptive alterations in

dopaminergic transmission by either disturbing or desensitizing the reward system, whose

activation by METH induces addiction, characterized by an obsessive drug seeking

behavior (Volkow et al., 2011). Table 1.5 summarizes the main biochemical alterations

induced by METH.

Page 65: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

65

Table 1.5 – Effects of METH

Effect References

Impairment of mitochondrial function (Schinder et al., 1996; Brown and Yamamoto, 2003; Wu et al., 2007)

Decrease in DAT activity and expression (Saunders et al., 2000; Volkow et al., 2001c; Barr et al., 2006; Riddle et al., 2006)

Decrease in dopamine D2 receptors (Volkow et al., 2001d)

Decrease in brain glucose metabolism under high doses

(Huang et al., 1999)

Changes in VMAT-2 activity and subcellular localization

(Riddle et al., 2006)

Ca2+ influx (Schinder et al., 1996)

Dopamine oxidation and ROS generation (Cadet and Brannock, 1998; Sulzer et al., 2005; Barr et al., 2006)

Release of monoaminergic neurotransmitters (Kuczenski et al., 1995; Sulzer et al., 2005)

Neuronal cell death (Davidson et al., 2001; Cadet et al., 2003)

Inhibition of MAO activity (Mantle et al., 1976)

Increase the expression of TH (Sulzer et al., 2005)

Lipid peroxidation and protein carbonyls generation (Yamamoto and Zhu, 1998; Wu et al., 2007; Alves et al., 2009)

Increase activity of GSH (Harold et al., 2000)

RNS generation (Imam et al., 2001b)

Inhibition of the ETC (Burrows et al., 2000; Virmani et al., 2004; Brown et al., 2005)

Activation of pro-apoptotic cascades (Imam et al., 2001a; Jayanthi et al., 2004; Koo et al., 2011)

Decrease activity serotonin and serotonin transporters

(Armstrong and Noguchi, 2004; Kish et al., 2009)

Increase of glutamate levels in several brain regions

(Mark et al., 2007; Kerdsan et al., 2012)

BBB disruption and increase in the permeability (Kiyatkin and Sharma, 2009; Ramirez et al., 2009)

Induction of pro- or anti-inflammatory mediators (Gonçalves et al., 2008; Pendyala et al., 2012)

Legend: BBB – Blood brain barrier; DAT – Dopamine transporter; ETC – Electron transport chain; GSH –

Glutathione; MAO – Monoamine oxidase; RNS – Reactive nitrogen species; ROS – Reactive oxygen species;

TH – Tyrosine hydroxylase; VMAT-2 – Vesicular monoamine transporter 2;

Exposure to METH was previously shown to provide an excellent model to mimic these

neurodegenerative processes, both in vivo and in vitro (Ferrucci et al., 2008a). In the

present study, METH is used in order to clarify the molecular mechanisms underlying the

Page 66: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

66

putative neuroprotective role of ALC. METH is known to affect both the mitochondrial

(Brown and Yamamoto, 2003) and the dopaminergic function (Barr et al., 2006) by

promoting the production of ROS. In addition, METH-induced activation of mitochondria-

mediated intrinsic cell death signaling events is known to be associated with the loss of

mitochondrial membrane potential, the release of apoptogenic factors and subsequent

activation of apoptotic cascades, creating a suitable model for exploring the mechanisms

of action of ALC. METH doses were established based on previous studies from Fornai

and collaborators (Fornai et al., 2004) for the in vitro assays and from groups of Bowyer

and Fukumura for the in vivo assays (Bowyer et al., 1994; Fukumura et al., 1998).

The present work has urged subsequently to previous observations from our group

demonstrating that pre-treatment with ALC confers effective neuroprotection against

MDMA-induced neurotoxicity, preventing mitochondrial oxidative damage, reducing

carbonyl formation, decreasing mtDNA deletion and improving the expression of

respiratory chain components (Alves et al., 2009). These results emphasize the benefits

from ALC administration in neurodegenerative disorders. However, as the underlying

molecular mechanisms of ALC action remain unclear, here we propose to study those

mechanisms that are associated to mitochondria bioenergetics and to dopamine

neurotransmission.

1.9. CELLULAR AND ANIMAL MODELS

In the present study, the experimental component was divided into two phases: before

using animal models, experiments were performed using a suitable dopaminergic cell line,

aiming to determine the role of ALC in mitochondrial bioenergetics, morphology and

functionality. The rat pheochromocytoma (PC12) cell line is well known for its

catecholamine production. PC12 cell lines are often used as an in vitro model to mimic

dopaminergic function and predict the neurotoxicity of a variety of compounds acting on

central DA neurons (Schlegel et al., 2004; Piga et al., 2005). This cell line is widely used

due the similarities with central dopaminergic neurons, easy handling and replication and

low cost maintenance. PC12 cells are able to produce DA (Greene and Tischler, 1976)

and contain DA receptors in their external membranes (Sampath et al., 1994) (figure

1.12). Comparing PC12 cells to dopaminergic central neurons, PC12 cells also express

TH, but have less DAT and no expression of VMAT-2 (Fornai et al., 2007). Dopamine

content in PC12 cells is 10-fold less than that of DA terminals, L-DOPA amount is very

Page 67: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

67

similar, but norepinephrine levels are very low, while epinephrine is not produced by these

cells. Moreover, storage mechanisms in PC12 cells are less efficient (Fornai et al., 2007).

Figure 1.12 – Schematic representation of PC12 cell s in comparison to dopaminergic nerve

terminal . Main metabolic steps are also depicted in the figure. [From (Fornai et al., 2007), with

permission].

In the present study, aiming to evaluate the putative role of ALC over the function of

neurotransmitter systems, we have exposed PC12 cells pre-treated with ALC to different

concentrations of METH, for 24h or 72h. Despite the differences between PC12 cells and

central dopaminergic neurons, PC12 cells possess unique dopaminergic features that

make them particularly sensitive to METH and thus, suitable for our purposes (Greene

and Tischler, 1976; Fornai et al., 2007).

In the second phase of the experimental procedures, young adult C57BL/6J male mice

were used. The option for this strain is related to the well characterized brain anatomy

(MacKenzie-Graham et al., 2004) as well as neurotoxicity profile of METH (O'Callaghan

and Miller, 1994).

Page 68: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 69: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

69

1.10. OBJECTIVES

Aging and neurodegenerative diseases have been associated to significant changes in

the brain bioenergetics, involving not only impairments in glucose metabolism and

neurotransmitter systems, but also increased oxidative damage due to less efficient

antioxidant systems. On one hand, there is extensive evidence in the literature regarding

the mitochondrial involvement in these processes. On the other hand, it is also reported

that ALC might improve deleterious effects induced by aging and neurodegeneration by

acting at the mitochondrial level and neurotransmitters modulation. This dissertation aims

to contribute for a better understanding of the mitochondria-related molecular mechanisms

underlying the action of ALC in the brain.

In order to fulfil this general goal, more specific objectives were outlined:

A. ALC action on glucose uptake and mitochondrial f unction

o Evaluate if the administration of ALC has an effect on brain glucose metabolism

through the in vivo evaluation of glucose brain uptake, using 18F-FDG.

o Explore the role of ALC in mitochondrial bioenergetics, using a dopaminergic cell

line (PC12 cells), exposed to METH, known to induce reductions on glucose

metabolism.

o Determine if ALC is able to protect cells from loss of mitochondrial membrane

potential and prevent the activation of the apoptotic cascade.

B. ALC action on the dopaminergic function

o Evaluate the role of ALC over the dopaminergic function in vitro, through the

quantification of intra and extracellular DA levels.

o Verify the presumed role of dopamine D2 receptor expression as a mechanism of

action of ALC’s ability to inhibit dopaminergic toxicity through DA receptor imaging

with the radiotracer 123I-Iodobenzamide.

Page 70: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 71: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

CHAPTER 2

ACETYL-L-CARNITINE IN MITOCHONDRIAL

BIOENERGETICS AND FUNCTIONALITY

Page 72: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 73: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

73

2.1. INTRODUCTION

Acetyl-L-Carnitine (ALC) is the most widely distributed ester of L-carnitine in the body,

playing a variety of vital functions. Due to its close interaction with cell bioenergetics, ALC

plays a role in metabolic compromising diseases, especially those related to the

mitochondria (Rosca et al., 2009). Although the role of ALC at the mitochondrial level has

been widely investigated, the molecular underlying mechanisms remain elusive.

It is currently thought that improving mitochondrial function and raising ATP levels may be

relevant for improving neuronal dysfunction (Owen and Sunram-Lea, 2011). In patients

with compromised cognitive function, ALC promoted an effective recovery of attention

capacity, mental flexibility, short-term memory, visual scanning and tracking, motor control

and orientation ability [revised by (Flanagan et al., 2010)]. We have demonstrated that

ALC confers effective neuroprotection against 3,4-methylenedioximethamphetamine

(MDMA)-induced neurotoxicity, preventing mitochondrial oxidative damage and the typical

MDMA-induced loss of serotonin (Alves et al., 2009). However, until now, the majority of

such studies were conducted in aged or demented populations with depleted energy

resources. Therefore, the benefits of ALC to healthy and young populations are unknown.

Besides the well-known role of ALC in fatty acid oxidation, it has been shown to exert

beneficial effects in patients with impaired fasting glucose and type 2 diabetes (Owen and

Sunram-Lea, 2011). Carnitine participates in the glucose metabolism by reducing the acyl-

CoA/CoA ratio in mitochondria, which in turn increases pyruvate dehydrogenase activity,

improving glucose disposal (Stephens et al., 2007). In addition, carnitine seems to have

beneficial effects through the modulation of insulin-like growth factors (IGFs) and IGF

binding proteins (Heo et al., 2001; Molfino et al., 2010) and by affecting the expression of

glycolytic and gluconeogenic enzymes (Hotta et al., 1996; Ruggenenti et al., 2009).

Recently, ALC was used in vitro to prevent METH-induced decrease of glucose uptake

and to stabilize the glucose transporters GLUT1 and GLUT3 (Abdul Muneer et al., 2011b).

In order to clarify the role of ALC in the neuronal metabolism of both healthy and

compromised subjects, we have exposed C57BL/6J mice to the action of a well known

dose of METH. We used positron emission tomography (PET) to evaluate changes in the

regional glucose uptake, which were paired to cognitive performances in a Water Maze

(WM). To further understand the mechanisms involved at the mitochondrial level, an in

vitro model of exposure to METH and ALC, was also used.

Page 74: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

74

2.2. MATERIALS AND METHODS

2.2.1. In Vivo Assessment of Glucose Uptake

Twelve week-old male C57BL/6J mice (21.6 – 26.0 g) were purchased from Charles River

(Innovo, Gödöllı, Hungary) and maintained on a 12-h light/dark cycle. All experimental

procedures were approved by the Local Animal Experimental Ethical Committee of

Semmelweis University, Budapest, Hungary, in compliance with the European Community

Council Directive of September 22, 2010 (2010/63/UE). All efforts were made to ensure

minimal animal stress and discomfort. Room temperature was kept at 22ºC and animals

were allowed water and food ad libitum except in the day of the study when they were

deprived of food 4h before 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) administration (free

access to water was maintained). FDG consists in a modified glucose molecule in which

the C-2 hydroxyl group was substituted by fluorine (18F). This modification allows FDG to

continue to be a substrate for the glucose transporter and for hexokinase conversion to

FDG-6-phosphate, but it cannot be metabolized beyond this first step. Consequently, 18F-

FDG is trapped in the cell as FDG-6-phosphate and its uptake can be used to determine

glucose uptake.Twelve animals were divided into four groups, according to different

treatments: group 1 (control, saline), group 2 (ALC, 100 mg/kg), group 3 (METH, 10

mg/kg) and group 4 (ALC + METH 30 min later). Drugs were administered by

intraperitoneal (ip) injection. Thirty minutes after the last dose, 10.5 ± 1.1 MBq of 18F-FDG

was injected in the tail vein, under anesthesia (isoflurane 2%). After injection, animals

recovered very quickly from isoflurane anesthesia and were returned to their home cage

for the 18F-FDG uptake period (45 minutes). Immediately before image acquisition,

animals were anaesthetized with urethane ip injection (1g/kg), the head was fixed to a

specific head holder bed (MultiCellTM, Mediso, Hungary) and the body immobilized with

tapes to prevent movement. Images were acquired in a PET-MRI scanner (nanoScan PM,

Mediso, Hungary), equipped with full ring LYSO crystals and one Tesla magnet. Images

were acquired for 50 minutes (25 minutes for each imaging modality).

List mode PET files were reconstructed into images of 3D volume of 0.3x0.3x0.3 mm

voxels by proprietary 3D ordered subset expectation maximization (OSEM) algorithm

(Tera-TomoTM, Mediso, Hungary) correcting for scatter. Decay-, random and dead time

correction was performed. A gamma photon energy window of 400 to 600 keV was used,

with 5 ns coincidence timing and 1:5 coincidence mode of the detector modules in the

ring. Regions of interest (ROIs) were drawn on several brain regions using the transverse

and coronal planes. For 18F-FDG brain uptake, the standardized uptake value (SUV) was

Page 75: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

75

calculated at each time point by dividing the mean radioactivity concentration (MBq/mL)

by the injected dose per body weight (MBq/g). For the MRI scans, a T1-weighted three-

dimensional Gradient Echo (GRE) sequence of the nanoScan PET-MRI scanner was

applied at the brain region, in 50 slices and 0.4 mm slice thickness. The frequency

direction being horizontal, the frequency/phase distribution was 192/192, with 5

excitations, 25 ns repetition time, 2.3 ns echo time and 25 degrees flip angle. All images

were co-registered and visualized using Interview Fusion TM (Mediso, Hungary) and

VivoQuant (inviCRO, Boston, USA) dedicated image processing software. We also used

advanced post processing Hybrid Viewer™, courtesy from HERMES Medical Solutions

(Stockholm, Sweden). Volume of interest delineation in 3D mode was done by fusing the

Brookhaven Laboratory Mouse MRI atlas developed at the University of Florida

(http://brainatlas.mbi.ufl.edu/) to the isotopic modality and matching the regional volumes

with the animal’s own MRI scans.

2.2.2. Assessment of Brain Igf1 and Igf1R Transcrip t Levels by Real Time

Polymerase Chain Reaction (RT-PCR)

Another set of 12 C57BL/6J mice were treated as described above and killed under

ketamine (75mg/kg)/medetomidine (1mg/kg) anesthesia. Brains were immediately

dissected on ice and brain regions frozen on dry ice cooled isopentane and stored at -

80ºC until use. For each brain region, total RNA was extracted using RNeasy® Mini Kit

(Qiagen, Hilden, Germany). RNA quality was confirmed by Experion automated

electrophoresis system (Bio-Rad Hercules, CA, USA). Synthesis of cDNA was performed

by Qiagen RT2 HT First Strand cDNA kit. A total amount of 2.0 µg of RNA was used to

study gene expression through quantitative RT-PCR.

2.2.3. Cell Culture and Drug Regimen

Rat pheochromocytoma PC12 cells (American Type Cell Culture Collection, Manassas,

VA) were used as an in vitro model to study neuroprotective effects of ALC. The cells

were used in their undifferentiated form between passage 12 and 16. Cells were grown in

Dulbecco’s Modified Eagle Medium (DMEM + GlutaMAX™-I, Gibco®) supplemented with

10% of fetal bovine serum (FBS, Gibco®) and 1% of penicillin (50 IU/mL) streptomycin (50

mg/mL) (PS, Gibco®) in a CO2 incubator at 37°C, with 5% CO 2 and 95% filtered air. Cells

Page 76: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

76

in log phase were treated with increasing doses of ALC (0.01, 0.1 and 1.0 mM) alone or in

combination with METH 1.0 or 100 µM, directly dissolved in low glucose (LG, 5.56 mM,

MEM-Alpha, Gibco®) or high glucose (HG, 25.0 mM, DMEM + GlutaMAX™-I, Gibco®)

concentration culture medium for 24h or 72h. When ALC and METH were combined, pre-

treatment with ALC preceded METH exposure in 30 min. The selected doses of METH

were previously described and well characterized (Fornai et al., 2004; Fornai et al., 2007).

ALC was kindly provided by Sigma-Tau, S.p.A. (Pomezia, Italy). METH hydrochloride was

obtained from Sigma-Aldrich® (St. Louis, MO). All subsequent experiments were

performed using culture medium without FBS in order to counteract the protective effect

conferred by the serum.

2.2.4. In Vitro Assessment of Glucose Uptake

Glucose uptake was measured through the quantification of the cellular uptake of 18F-

FDG. 8x105 cells/mL were seeded in 12-well plates 24h prior to treatment with ALC and

METH. Cells were then incubated with 37 kBq/mL of 18F-FDG in a low glucose serum-free

medium for 60 min. Cells were lysed by adding NaOH (1.0 M), assayed for radioactivity by

gamma-counting (Berthold LB 2111 gamma counter) and the total protein content was

determined (Aloj et al., 1999; Smith, 2001).

2.2.5. Cell Viability Assay

To assess cell viability the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

(MTT) assay was used. This is a standard colorimetric assay for measuring the activity of

enzymes that reduce MTT to formazan, giving a purple color. This reaction takes place

only when mitochondrial reductase enzymes are active, and therefore conversion can be

directly related to the number of viable (living) cells (Mosmann, 1983; Ahmadian et al.,

2009). Cells were seeded in 24-well plates at a density of 8x104 cells/mL. At cell

confluence, drugs were added and cultures incubated for 24 or 72h either in LG and HG

culture medium. Then, medium was discarded and replaced by 100 µL of fresh medium

containing 1.0 mM of MTT (Sigma Chemical Co., St. Louis, MO) and incubated for 3h at

37°C in the CO2 incubator. Medium was discarded and 100 µL of dimethyl sulfoxide

(DMSO, Sigma Chemical Co., St. Louis, MO) was added to dissolve the produced

formazan crystals. Absorbance at wavelength 540 nm was measured in a microplate

reader.

Page 77: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

77

2.2.6. Determination of Mitochondrial Membrane Inte grity

MitoTracker®Red CMXros (Molecular Probes) was used to assess mitochondrial

membrane integrity; this mitochondria-selective probe, passively diffuses across the

plasma membrane and accumulates in active mitochondria. Procedures were carried out

according to manufacturer’s instructions. Briefly, cells were seeded in 24-well plates at a

density of 3x105 cells/mL. 24 hours later, cells were treated with the above mentioned

concentrations of ALC and METH and incubated for additional 24h or 72h. Cells were

then stained with 1.0 µM of MitoTracker® Red CMXros and incubated at 37°C for 30 min

in the dark. Cells were washed twice, harvested and centrifuged. Cells were then

transferred to a tube on ice for analysis of the fluorescence intensity by flow cytometry

(FACS CaliburTM, BD Biosciences). Fluorescence images were also acquired in confocal

microscope Leica TCS SP5 II.

2.2.7. Assessment of ATP Synthesis

ATP synthesis was evaluated by luminescence using the kit CellTiter-Glo® (Promega),

which signals metabolically active cells through the quantitation of the ATP present. Cells

were seeded in 24-well plates at a density of 4x104 cells/mL. Drugs were added 24 hours

later and incubated for additional 24h or 72h in LG or HG concentration culture medium.

CellTiter-Glo® reagent was added to each well in an equal volume to the cell culture

medium. Cells were transferred to 96-well opaque-walled plates and readings were

performed in the plate reader (BioTek Synergy 2) with an integration time of 1

second/well. Control wells (medium without cells) were prepared to determine background

luminescence. A standard curve per assay was also generated. In order to study the

origin of the synthesized ATP (glycolysis or oxidative phosphorilation), an inibitor of the

complex III of the electron transport chain (antimycin A) was also added in some

experiments. Antimycin A (4.0 µM) was added to culture medium 30 minutes before ATP

assessment.

2.2.8. Assessment of Mitochondrial Mass

MitoTracker® Green FM (Molecular Probes) was used to quantify mitochondrial mass; this

mitochondria-selective probe, passively diffuses across the plasma membrane and binds

to lipids from the inner mitochondrial membrane (Lee et al., 2000). Procedures were

Page 78: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

78

carried out according to manufacturer’s instructions. Briefly, cells were seeded in 24-well

plates at a density of 3x105 cells/mL. 24 hours later, cells were treated with the above

mentioned concentrations of ALC and METH and incubated for additional 24h or 72h.

Cells were then stained with 800 nM of MitoTracker® Green FM and incubated at 37°C for

30 min in the dark. Cells were washed twice, harvested and centrifuged. Cells were then

transferred to a tube on ice for analysis of the fluorescence intensity by flow cytometry

(FACS CaliburTM, BD Biosciences).

2.2.9. Determination of Mitochondrial Membrane Pote ntial ( ∆ψm)

Mitochondrial membrane potential was measured using the radioactive lipophilic and

cationic agent, hexakis (2-methoxyisobutylisonitrile)-technetium-99m (99mTc-sestamibi),

initially developed for myocardial perfusion imaging. The uptake of this agent depends on

mitochondrial metabolism as well as negative potential of the inner membrane (Piwnica-

Worms et al., 1990; Moretti et al., 2005). Briefly, 25 x104 cells were seeded in 12-well

plates 24h prior to treatment with ALC and METH. Cells were then incubated with 74

kBq/mL of 99mTc-sestamibi for 45 min. Cells were lysed by adding NaOH (1.0 M), assayed

for radioactivity by gamma-counting (Berthold LB 2111 gamma counter) and total protein

content was determined.

2.2.10. Determination of Bax Activation and Locatio n

The expression of Bax protein was monitored by western blot, using two different

antibodies, one for active Bax (6A7, abcam) and another one for inactive form (Cell

Signaling Technology). Cells were seeded in 100 mm tissue culture dishes (Orange

Scientific) at a density of 4x104 cells/mL. At 80% of confluence, ALC 0.01 mM alone or in

combination with METH 1.0 µM and 100 µM was added and cells were incubated for 24h.

Cells were then lysed in TEN buffer (50 mM Tris-HCl, 0.2 M EDTA, 150 mM NaCl, 1%

NP-40, 4.5 mM sodium pyrophosphate, 10 mM ß-glycerophosphate, 1.0 mM sodium

fluoride, 1.0 mM sodium orthovanadate and proteases inhibitors 1:500, Sigma, Missouri,

USA) and proteins were extracted. The supernatant was taken for western blot analysis,

40 µg of protein was separated on 10% SDS-polyacrylamide gels. Proteins were

transferred to a polyvinylidene fluoride membrane for blotting (0.2 µm, Bio-Rad) by

electroblotting (Trans-Blot Turbo Transfer System for Protein, Bio-Rad). The membranes

were blocked in 3% BSA (Nzytech) in 0.1% TBS Tween 20 (50 mM Tris, 150 mM NaCl,

Page 79: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

79

pH 7.6), 1h at room temperature. Then, were incubated with primary antibodies, active

anti-Bax (0.5 µg/mL) and and anti-GAPDH (1:200000) (5G4, HyTest) diluted in 3% BSA in

0.1% TBS-T, overnight at 4ºC. Membranes were washed three times for 10 min in the

same buffer and incubated for 1h at room temperature with Immunopure Goat anti-mouse

IgG peroxidase conjugated (1:1000) (Thermo Scientific) and anti-rabbit IgG peroxidase

conjugate (1:1000) (Sigma).

Immunoreactive proteins were revealed using enhanced chemiluminescence method

(Immun-Star HRP Chemiluminescent kit, Bio-Rad). The membranes were washed and

incubated in a Restore Western Blot Stripping Buffer (Thermo Scientific), for 15 min at

room temperature. Then the membranes were blocked in 3% BSA in 0.1% TBS-T, 1h at

room temperature and were incubated with primary antibodies inactive anti-Bax (1:1000,

Cell Signaling) and anti-GAPDH, overnight at 4ºC. The blots were probed with horseradish

peroxidase-labeled secondary antibodies. Blots were analyzed with Quantity One 1-D

Analysis Software, version 4.6 (Bio-Rad). Additionally, Bax location was monitored by

confocal microscopy. Briefly, cells were seeded in 24-well plates over glass coverslips

treated with poly-L-lysine at a density of 3x105 cells/mL. 24 hours later, cells were treated

with ALC 0.01 mM and METH and incubated for additional 24h. Cells were then washed

and 500 nM of MitoTracker CMXRos was added for 30 min at 37ºC in the dark. 4%

formaldehyde was used for cell fixation during 20 minutes after which cells were

permeabilized with 0.2% Triton for 2 minutes and blocked for 1h with 5% BSA. Cells then

incubated with anti-Bax antibody (5ug/ml, diluted in 5% BSA) overnight at 4ºC. Cells were

washed and the secondary antibody was added (anti-mouse 1:1000, Alexa Fluor 488)

followed by 1h of incubation. Cells were washed four times and coverslips were mounted

directly in aqueous mounting medium with DAPI (1:10 000, Sigma). Immunofluorescence

images were acquired in confocal microscope Leica TCS SP5 II.

2.2.11. Statistical Analysis

All data are representative of at least three independent experiments. Data are presented

as mean+SEM. Statistical significance was determined by using one-way ANOVA with

Bonferoni’s post hoc test. 18F-FDG-PET data were analyzed by two-way ANOVA with

Bonferoni’s post hoc test. Differences were considered to be statistically significant at

p<0.05 level. All analyses were performed using Prism 6 for Mac OS X (GraphPad

Software, San Diego, California, USA).

Page 80: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

80

2.3. RESULTS

2.3.1. ALC Preconditioning Increased Glucose Uptake in METH Exposed

Animals

We have used a C57BL/6J mice model to evaluate the role of ALC in the cerebral glucose

metabolism. We have used a well-described dose of METH to clarify the role of ALC

under glucose-deprived conditions. Our analysis was focused in regions receiving relevant

dopaminergic inputs (and thus preferentially affected by METH), such as the prefrontal

cortex (PFC), the striatum (left and right) and the hippocampus. Due to its strong

regulation by GABAergic striatal inputs, the thalamic region was also included. The dose

of ALC administered (100 mg/kg) was previously shown to modulate glucose metabolism

in the brain (Scafidi et al., 2010a). Furthermore, we have previously demonstrated that

this ALC dose was effective in preventing the toxic effects of MDMA at the mitochondrial

level (Alves et al., 2009). Interestingly, our present results show that neither ALC nor

METH had a significant impact on glucose uptake in none of the assessed brain regions.

However, when ALC is administered 30 min before METH, glucose uptake is highly

augmented (two-way ANOVA (treatment x region), F(12,40)=0.5408, p<0.0001 for

treatment, p=0.0016 for brain region, and no interaction between treatment and region).

The PFC was one of the most affected regions, displaying an increased uptake in the

ALC/METH group that was highly significant when compared to all other conditions

(p<0.00001 to ALC and METH and p=0.0228 to control, Bonferroni’s post-hoc, figure 2.1-

A and B). The right striatum was also highly affected, displaying a very similar increase in

the ALC/METH group (p<0.00001 to ALC and p=0.0002 to METH and p=0.0097 to

control, Bonferroni’s post-hoc, figure 2.1-A and B). In the left striatum, differences were

significant when compared to ALC (p=0.0006, figure 2.1-A and B) and METH (p=0.0083,

figure 2.1-A and B), but not to control. Likewise, the thalamic region displayed an

increased uptake for the ALC/METH group when compared to ALC (p=0.0008, figure 2.1-

A and B) and METH (p=0.0353, figure 2.1-A and B). The uptake in the hippocampus was

less affected by ALC/METH and presented only an augmented uptake when compared to

ALC (p=0.0065, figure 2.1-A and B). Of note, the intense uptake characteristic of the

tongue and salivary glands, particularly in the presence of an adrenergic agent such as

METH, is no longer observed. Likewise, the uptake in the harderian glands seems to be

clearly reduced, however these extra cerebral regions were not quantified.

Page 81: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

81

Figure 2.1 - Increased glucose uptake in the brain after the administration of METH to mice

preconditioned with ALC . A: Representative 18F-FDG-PET-MRI images from 12-week old male

mice from different groups: control, ALC, METH and ALC/METH. In the upper row, representation

of the regions of interest (ROI) used for SUV quantification. Tomographic images were

reconstructed by OSEM algorithm and the resultant planes are shown: sagittal, transverse and

coronal. Note the generalized increase in glucose uptake on ALC/METH group. Extra cerebral

intense 18F-FDG uptake corresponds to a characteristic pattern of harderian glands (very well

depicted in the control group) and to the tongue and salivary glands, particularly in the presence of

METH. B: Bar graph representing SUV quantification of brain glucose uptake using 18F-FDG-PET.

Glucose uptake was assessed in the prefrontal cortex (PFC), left and right striatum (left and right

striat), hippocampus (hippoc) and in the thalamus. Data represent mean+SEM (n=3 mice/group).

SUV is consistently higher in ALC/METH group comparing to ALC in all brain regions. Significant

differences are marked as **p<0.01 as compared with the control condition; �p<0.05, ��p< 0.01and

���p< 0.001 as compared with METH and �p<0.05, ��p<0.01 and ���p<0.001 as compared

with ALC (as determined by two-way ANOVA, followed by a post-hoc Bonferroni’s test).

2.3.2. Igf1 and Igf1R Regulation

IGF1 promotes glucose utilization and is known to be strongly expressed in response to

several types of brain insults (Gehrmann et al., 1994; Jin et al., 2001; Fernandez and

Torres-Aleman, 2012). Additionally, carnitine supplementation was seen to activate the

Page 82: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

82

IGF1 signaling cascade (Heo et al., 2001; Bondy and Cheng, 2004) and increase the

plasmatic levels of IGF1 (Heo et al., 2001). On the other hand, METH seems to decrease

IGF1 receptors levels (Bourque et al., 2011). Therefore, we investigated the transcript

levels of Igf1 and the Igf1R in the different experimental conditions used in our in vivo

model, measured 24h after the drug administration period. This evaluation was conducted

in the striatum, which is known to be particularly susceptible to METH. Our results

evidenced that exposure to METH induced a significant decrease in Igf1 levels in the

striatum (one-way ANOVA, F(3,12)=28.700, p=0.0007, Bonferroni’s post-hoc, figure 2.2-

A). Importantly, the combined administration of ALC and METH prevented the decrease in

Igf1 induced by METH (p=0.0010). ALC administration by itself increased Igf1 expression

(p=0.0206, figure 2.2-A), which is in accordance with previous reports (Ahmed, 2012) and

seems to be relevant for the putative neuroprotective features of ALC. Therefore, Igf1

levels were affected within the experimental groups (ALC and METH) that did not show a

significant decrease in glucose uptake, but unaltered in the ALC/METH that displayed

highly increased glucose uptake. Of note, transcript levels of Igf1R were significantly

increased in the ALC/METH group (one-way ANOVA, F(3,11)=17.23, p=0.0032 to control,

p=0.0001 to METH and p=0.0104 to ALC Bonferroni’s post-hoc, figure 2.2-B).

Figure 2.2 - Igf1 (A) and Igf1R (B) transcript leve ls in the striatum of mice from different

groups (n=3 mice/group). Bar graphs represent the relative fold change to the control condition.

Note that ALC increases Igf1 levels, while METH decreases it. Animals preconditioned with ALC

and exposed to METH showed increased levels of Igf1R when compared to all other groups.

Significant differences are marked as *p<0.05, **p<0.01 and ***p<0.001, as compared with the

control condition; ���p< 0.001 as compared with METH and �p<0.05 as compared with ALC (as

determined by one-way ANOVA, followed by a post-hoc Bonferroni’s test).

Page 83: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

83

2.3.3. ALC Affects Glucose Uptake in a Dose-depende nt Way

To clarify the action of ALC in the metabolism of neuronal cells, we have also assayed the

uptake of glucose in the PC12 dopaminergic cell line. Despite the differences between

PC12 cells and the central dopaminergic neurons, PC12 cells possess unique

dopaminergic features that make them particularly sensitive to METH (Greene and

Tischler, 1976; Fornai et al., 2007) and, therefore, suitable for our purposes. The assay is

performed necessarily in cells kept in a LG medium. Consistently with what we observed

in vivo, METH did not show an effect on glucose uptake. ALC however, seems to have a

dose-dependent effect on the uptake of glucose, since at 24h ALC 0.01 mM, by itself,

significantly increased the uptake of glucose (one-way ANOVA, F(3,22)=3.432, p=0.0352,

Bonferroni’s post-hoc, figure 2.3-A). Likewise, the combined action of ALC 0.01 mM and

METH 1.0 or 100 µM, induced an increased glucose uptake (one-way ANOVA,

F(4,21)=3.256, p=0.0368 for ALC/METH1 and F(4,30)=9.232, p=0.0004 for

ALC/METH100, Bonferroni’s post-hoc, figure 2.3-B and C) but at 72h this effect was no

longer seen. For METH 1.0 µM, at 72h, the combined action of ALC 0.01 mM was even

lower than in control conditions (one-way ANOVA, F(4,45)=24.430, p=0.0015 to control

and p<0.0001 to METH1, Bonferroni’s post-hoc, figure 2.3-E). ALC 1.0 mM, however, lead

to increased uptake at 72h, either by itself (one-way ANOVA, F(3,36)=26.510, p<0.0001,

Bonferroni’s post-hoc, figure 2.3-D), or in the presence of METH1 (one-way ANOVA,

p<0.0001 to control and p=0.0015 to METH1, Bonferroni’s post-hoc, figure 2.3-E) and

METH100 (one-way ANOVA, F(4,47)=4.005, p=0.0117, Bonferroni’s post-hoc, figure 2.3-

F). The selected doses of METH were based on previous dose-response studies (Fornai

et al., 2007) demonstrating that an intraperitoneal dose of 5 mg/kg of this compound

results in a striatal dose of 1.0 µM, while a 100 µM dose is much more toxic. The absolute

glucose uptake by control cells was very similar at 24h and 72h (data not shown).

Since the ALC 0.01 mM dose seems to have more impact on the short term glucose

uptake, we selected this dose to proceed with the in vitro evaluation.

Page 84: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

84

Figure 2.3 - Altered glucose uptake in long time st arvation in PC12 cells exposed to ALC . A

to C: Cells treated with ALC at 0.01, 0.1, 1.0 mM alone or in combination with METH 1.0 µM or 100

µM for 24h. The lowest ALC dose increases glucose uptake at 24h, alone (p=0.0352) or in

combination with METH 1.0 and 100 µM (p=0.0004). D to F: Cells treated with ALC at 0.01, 0.1, 1.0

mM alone or in combination with METH 1.0 µM or 100 µM for 72h. In conditions of long time

starvation, ALC seems to have a dose-dependent effect, increasing significantly glucose uptake

with the ALC dose (p<0.0001 for ALC 1.0 mM comparing to control). Graphic representation of

glucose uptake per mg of total protein content, comparing to control cells. Columns represent

mean+SEM (n=9). Significant differences are marked as *p<0.05, **p<0.01 and ***p<0.001, as

compared with the control condition and �p<0.05, ��p<0.01 and ���p<0.001 as compared with

METH 1.0 or 100 µM (as determined by one-way ANOVA followed by a post-hoc Bonferroni’s test).

Dashed lines represent the control condition.

2.3.4. ALC/METH Prevented the METH-induced Decrease in Mitochondrial

Function

To better characterize the action of the selected ALC dose, we have performed an MTT

assay, which measures the activity of dehydrogenase enzymes, in metabolically active

mitochondria. This assay reflects both cell viability and the enzymatic functionality of

mitochondria. We evaluated the action of ALC in undifferentiated PC12 cells cultured in a

Page 85: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

85

low glucose environment. At 24h, the combination of ALC 0.01 mM and METH resulted in

a significant decrease in MTT reduction (one-way ANOVA, F(5,145)=9.701, p<0.0001 for

ALC0.01/METH1 and for ALC0.01/METH100, Bonferroni’s post-hoc, figure 2.4-A)

reflecting decreased cellular viability. METH by itself was not different from the control. At

72h the decrease in MTT reduction was still present for ALC0.01/METH1 (one-way

ANOVA, F(5,130)=7.096, p=0.0002, Bonferroni’s post-hoc, figure 2.4-B). At 72 h, METH

100 µM also led to lower levels of MTT reduction (p=0.0188) (figure 2.4-B), and therefore

at this time point ALC may have contributed to balance the effect of METH, since the

combined action of METH100 and ALC 0.01 mM is not different from the control.

Figure 2.4 – Cell viability of PC12 cells at 24 and 72h. A: Decrease in cell viability when ALC

0.01 mM was used in combination with METH1 or METH 100 (p<0.0001 for both). B: Cell viability

in PC12 cells treated for 72h. Note the significant decrease in cell viability when cells were treated

with ALC 0.01/METH1 (p=0.0002) and METH100 (p=0.0188). In the latter case, ALC 0.01 mM was

able to prevent the decline in mitochondrial function induced by METH100. Significant differences

are marked as *p<0.05 and ***p<0.001, as compared with the control condition and ��p<0.01 and

���p<0.001 as compared with METH 1.0 or 100 µM (as determined by one-way ANOVA followed

by a post-hoc Bonferroni’s test). Dashed lines represent the control condition.

Mitochondrial function was assessed with the MitoTracker®CMXRos, a dye retained by

active mitochondria that are effectively contributing to cell respiration. At 24h, we did not

observe any significant differences between conditions (data not shown). However, for

prolonged exposures, METH significantly reduced mitochondrial function (both METH 1.0

Page 86: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

86

and 100 µM: one-way ANOVA, F(5,67)=11.270, p<0.0001, Bonferroni’s post-hoc, figure

2.5-A, C and D). This reduction was also visible in confocal microscopy (figure 2.5-E and

F). Importantly, in this case, the ALC dose 0.01 mM was effective in preventing the

reduction induced by METH 1.0 µM (one-way ANOVA, F(5,67)=11.270, p=0.0002,

Bonferroni’s post-hoc, figure 2.5-A, C and G).

Figure 2.5 - Decreased mitochondrial function in co nditions of long time starvation, induced

by METH . A: Mitochondrial function at 72h measured by flow cytometry using the MitoTracker®

CMXRos. In long time starvation, METH induced a near 50% reduction in mitochondrial function,

partially prevented by ALC 0.01 mM (for METH1). Graphic representation of the percent of cells

contributing to cell respiration comparing to control. Columns represent mean+SEM (n=9).

Significant differences are marked as *p<0.05 and ***p<0.001, when compared with the control

condition and ��p<0.01 and ���p<0.001 when compared with METH (as determined by one-way

ANOVA followed by a post-hoc Bonferroni’s test). Dashed lines represent control conditions. B to

D: representative fluorescence histograms of the flow cytometry quantification presented in A.

Graphics represent the range of red fluorescence and the number of cells associated to that

emission. E to G: Fluorescence images of PC12 cells stained with 150 nM of MitoTracker®

CMXRos (red staining) depicting mitochondrial function, and 4 µM of DAPI depicting cell nucleus

(blue staining). E, control; F, METH 1.0 µM; G, ALC 0.01 mM/METH 1.0 µM. Scale bar = 10 µm.

2.3.5. ALC/METH Decreased ATP Synthesis in Glucose Deprived Cells

Reduced levels of mitochondrial function should result in reduced ATP levels and

therefore we have measured the amount of ATP being synthesized within the different

Page 87: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

87

experimental conditions. In a LG medium, we have observed that the combined action of

ALC and METH resulted in lower ATP levels (one-way ANOVA, F(5,86)=7.597, p=0.0040

for ALC0.01/METH1 and p=0.0088 for ALC0.01/METH100, Bonferroni’s post-hoc, figure

2.6-A). Of note, this early reduction of ATP synthesis may be protective in the long term.

To verify if the amount of glucose in the medium was affecting the synthesis of ATP, we

have repeated the whole evaluation in a HG medium. Although in control conditions the

absolute production of ATP had always been higher in LG than in HG medium (p=0.0411

at 24h and p=0.0017 at 72h, figure 2.6-C), in the presence of ALC, even when combined

with METH, the ATP production is much higher in HG (one-way ANOVA, F(5,86)=6.011,

p<0.0001 for ALC 0.01 mM and p=0.0062 for ALC0.01/METH1, Bonferroni’s post-hoc,

figure 2.6-B). Interestingly, although mitochondrial function was more affected at 72h than

at 24h, the decrease in ATP synthesis, under the action of ALC and METH, seems to

precede it. Of note, the absolute amount of ATP produced by control cells at 72h was 25%

higher than at 24h (one-way ANOVA, F(3,68)=10.980, p=0.0386, Bonferroni’s post-hoc

figure 2.6-C). To verify if in HG conditions the extra amount of ATP is provided by the

activity of the respiratory chain, we added antimycin A to the culture medium to block

complex III activity. In the presence of antimycin A the amount of ATP produced was

similar to the control cells in all tested conditions (data not shown). Although PC12 cells

are known to be relatively resistant to antimycin A (Ohsawa et al., 2003; Han and Im,

2008), our results show that the absolute amount of ATP synthesized in the presence of

ALC alone or in combination with METH was significantly reduced when antimycin was

added, showing that the increase in ATP production in HG conditions is due to the ALC

effect over the respiratory chain activity.

Page 88: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

88

Figure 2.6 – ATP synthesis of PC12 cells under diff erent conditions . A: Relative ATP

synthesis of cells treated for 24h in LG medium expressed as a percentage of control. Note the

decreased ATP synthesis induced by the combined use of ALC 0.01 mM and METH1 (p=0.0004)

or METH100 (p=0.0088). B: Relative ATP synthesis of cells treated for 24h in HG medium,

expressed as a percentage of control. Note the increased ATP synthesis promoted by ALC 0.01

mM in HG medium. C: Absolute ATP synthesis of control cells at 24h and 72h in low and high

glucose culture medium. Control cells in LG medium exhibit a considerably higher ATP synthesis

than those cultured in HG medium (p=0.0411 for LG-24h vs. HG-24h and p=0.0017 for LG-72h vs.

HG-72h). Moreover, in LG medium, ATP synthesis was higher at 72h (p=0.0386). Columns

represent mean+SEM (n=14-18). Significant differences are marked as *p<0.05, **p<0.01 and

***p<0.001 as compared with the control condition and �p<0.05 and ���p<0.001 when compared to

the respective concentration of METH (as determined by one-way ANOVA followed by a post-hoc

Bonferroni’s test). Dashed lines represent control conditions.

2.3.6. Increased ROS Formation Associated Transloca tion of Active Bax to

the Nucleus

The production of intracellular ROS is a hallmark of METH action; therefore, we evaluated

the formation of ROS in our experimental model. Although ALC is often referred as an

antioxidant compound (Calabrese et al., 2006; Rump et al., 2010), we verified that in the

presence of both METH 1.0 and 100 µM, ALC 0.01 mM augmented the formation of ROS

when compared to control (one-way ANOVA, F(5,37)=2.591, p=0.0136 for

ALC0.01/METH1 and p=0.0193 for ALC0.01/METH100, Bonferroni’s post-hoc, figure 2.7-

A). Importantly, ROS production is in accordance with the results obtained for the ATP

synthesis. As ROS production might also influence mitochondrial membrane potential, we

questioned if the membrane potential would be compromised as well. To verify that, we

used a radioactive lipophilic and cationic agent, 99mTc-sestamibi, whose uptake is

dependent on the electropotential gradient across the mitochondrial membrane. We

observed a significant reduction in the membrane potential at 24h in cells precondioned

with ALC and exposed to METH 1.0 µM (one-way ANOVA, F(5,45)=5.159;

ALC0.01/METH1 p=0.0100 to control and p=0.0004 to METH1, Bonferroni’s post-hoc,

figure 2.7-B). Importantly, reduced membrane potencial is inversely related to ROS

production, which is in accordance with previous reports (Kim et al., 2004).

Page 89: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

89

Figure 2.7 - Decreased membrane potential in cells preconditioned with ALC and exposed to

METH. A: Intracellular ROS produced by cells treated in LG medium for 24h, expressed as mean

fluorescence intensity (MFI) per mg of protein (percent of control condition, n=9). Note the increase

in intracellular ROS production in the presence of ALC0.01/METH1 (p=0.0136) and

ALC0.01/METH100 (p=0.0193). B: Mitochondrial membrane potential of cells treated in LG

medium for 24h, expressed as percent uptake of 99mTc-sestamibi per unit of mitochondrial mass

(n=9). The combined use of ALC and METH significantly reduced the mitochondrial membrane

potential, particularly for ALC0.01/METH1 (p=0.0100 to the control p=0.0004 to METH1).

Significant differences are marked as *p<0.05 as compared with the control condition and

���p<0.001 when compared to METH1 (as determined by one-way ANOVA followed by a post-hoc

Bonferroni’s test). Dashed lines represent control conditions.

As a consequence of increased ROS, Bax, a protein involved in the regulation of apoptotic

cascade (Hsu and Youle, 1997), can be activated leading to increased mitochondrial

transition pore formation (Lee et al., 2012), associated with loss of mitochondrial

membrane potential. When evaluating the presence of activated Bax, we found no

significant differences between groups (figure 2.8-B). However, immunocitochemistry of

activated Bax, revealed interesting differences in its redistribution. Of note, while METH

seems to reduce its translocation to the mitochondria (figure 2.8-A), ALC seems to

promote its translocation to the nucleus. Moreover, when ALC was combined with METH,

Bax is seen in the nucleus, but also in the mitochondria. These observations match

previous reports, showing Bax translocation from the cytosol to the mitochondria, nucleus

and endoplasmic reticulum after the exposure to cytotoxic agents (Gajkowska et al.,

2001).

Page 90: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

90

Figure 2.8 – Translocation of active Bax to the nuc leus . A: Immunofluorescence images of cells

treated in LG medium for 24h and stained with DAPI (blue), 6A7 antibody for active Bax (green)

and MitoTracker® CMXRos (red). Note the translocation of Bax to the nucleus when cells were

treated with ALC0.01/METH1. Scale bar = 10 µm. B: Ratio of active to total Bax, expressed as a

percentage of the control condition. Columns represent mean+SEM (n=3).

Furthermore, mitochondrial mass was evaluated using the lipophilic and cationic

properties of Mitotracker® Green FM that accumulates preferentially in the mitochondria,

binding to the lipids from the inner mitochondrial membrane, independently of the

mitochondrial membrane potential (Lee et al., 2000; Pendergrass et al., 2004). Data

obtained for mitochondrial mass at 24h revealed decreased mass for ALC0.01/METH100

(one-way ANOVA, F(5,58)=7.475, p=0.0034, Bonferroni’s post-hoc), but increased for

METH100 (p=0.0248) (figure 2.9-A). At 72h, both METH100 and ALC0.01/METH led to

lower mitochondrial mass (one-way ANOVA, F(5,78)=5,307, p<0.0001 for METH100 and

p=0.0170 ALC0.01/METH100, Bonferroni’s post-hoc, figure 2.9-B).

Page 91: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

91

Figure 2.9 – The combined use of ALC and METH decre ases mitochondrial mass . This effect

is seen both at 24h (A) and 72h (B). Of note is the early transitory increase in mitochondrial mass

after exposure to METH 100 µM, which drops significantly after a prolonged exposure. Significant

differences are marked as *p<0.05, **p<0.01 and ***p<0.001 as compared with the control

condition and ���p<0.001 when compared to METH100 (as determined by one-way ANOVA

followed by a post-hoc Bonferroni’s test). Dashed lines represent control conditions.

2.4. DISCUSSION AND CONCLUSION

The present study unravels potential deleterious effects of ALC supplementation to

healthy or metabolically compromised individuals. In mice preconditioned with ALC and

exposed to METH, we verified a generalized increase in glucose uptake in several brain

regions, which was particularly evident in regions receiving a strong dopaminergic input.

Importantly, these animals performed poorly in a water maze test (performed in the lab,

but not addressed in the present work), reflecting impaired reference memory. In vitro, this

was associated with higher levels of intracellular ROS, decreased mitochondrial

membrane potential and translocation of active Bax to the nucleus.

In human METH abusers glucose metabolism is persistently reduced in the striatum, the

hippocampus or the thalamic region (Volkow et al., 2001a; Wang et al., 2004; London et

al., 2005). However, when assessing the effect of a single low dose of METH in naïve

mice, we verified that glucose uptake was not affected in any of the analyzed regions.

Since the acute effect of a single low dose is not comparable to a chronic exposure, this is

not unexpected. Importantly, in animals preconditioned with ALC, the administration of

Page 92: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

92

METH led to a robust and general increase of glucose metabolism in the whole brain,

which was particularly relevant in the PFC.

Recently, using 13C-ALC, it was demonstrated that the acetyl moiety of ALC both in

astrocytes and neurons is rapidly incorporated into glutamate and GABA synthesis

(Scafidi et al., 2010b). This establishes the direct involvement of ALC in increased

glutamine, glutamate and GABA levels, via the tricarboxilic acid (TCA) cycle. Importantly,

biosynthesis of both tyrosine and L-tryptophan, dopamine and 5-HT precursors, involves

either a transamination reaction with glutamate or the participation of glutamine as an

amine donor (Yudkoff, 1997), which may account to explain the action of ALC over

monoamine production.

In astrocytes, the increased routing of glutamate into the TCA cycle seems to rise ATP

levels, leading to phosphofructokinase and glycolysis inhibition, and promoting glyocogen

synthesis [reviewed by (DiNuzzo et al., 2011)]. On the other hand, stress events, such as

METH exposure, would favor glycogen degradation (Cruz and Dienel, 2002). In vivo,

although glutamatergic neurotransmission accounts for nearly 85% of intracortical

signaling, it seems that in regions receiving stronger noradrenergic and serotonergic

inputs, the activity of these neurotransmitters may be crucial in determining the amount of

glycogen that will be mobilized (DiNuzzo et al., 2011). Therefore, the combined action of

METH over dopaminergic and adrenergic receptors and transporters, allied to ALC-

induced synthesis of neurotransmitters may justify the increase of glucose uptake that we

have observed.

It has been suggested that glucose uptake is influenced by the expression of IGF1

through the indirect promotion of GLUTs (1, 3 and 4) translocation to the plasma

membrane, thus augmenting glucose transport into the cell (Bondy and Cheng, 2004).

Because the brain relies essentially on glucose oxidation for energy production, insulin

and IGF receptors are highly expressed in this organ, particularly in neurons and, to a

lesser extent, in glia and other areas (Gerozissis et al., 2001). Within the present work, we

observed that METH exposure reduced Igf1 levels, while ALC enhanced it. Of note, ALC

preconditioning in METH exposed animals led to Igf1 levels that were similar to the

control. Increasing IGF1 expression may be an attempt to promote GLUTs translocation to

the outer cell membrane, thus augmenting glucose uptake by the brain, which seems to

be relevant for the action of ALC. In accordance, increased IGF1 expression has been

reported in response to several types of injury (Genead et al., 2012; Schober et al., 2012).

While it is possible that increased levels of IGF1 in the brain may constitute a

compensatory mechanism to prevent metabolism disruption, it was also suggested that

Page 93: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

93

IGF1 may not be directly involved in the uptake of glucose by neurons (Tafreshi et al.,

2010). Interestingly, we verified that the levels of Igf1R were only upregulated for the

ALC/METH group, where Igf1 was unaltered, but glucose uptake was highly increased.

Augmented Igf1R levels were observed in the brain of rats kept under a low carbohydrate

diet (Cheng et al., 2003). Of note, it was demonstrated that IGF1 uptake through the blood

brain barrier is modulated by neural activity (Nishijima et al., 2010) and therefore,

unaltered Igf1 levels do not necessarily correspond to unaltered IGF1 levels.

To better understand the role of ALC in neuronal glucose uptake under the action of

METH, we have used a dopamine producing cell line that was exposed to METH and ALC

in a glucose-deprived condition. In these cells, an ALC dose of 0.01 mM led to improved

glucose uptake alone or in combination with METH, which was followed by a strong

decrease at 72h. In a previous work, in a primary neuronal culture, reduced neuronal

glucose uptake after METH (100µM) was paralleled by lower expression of GLUT3 (Abdul

Muneer et al., 2011b). Interestingly, these authors demonstrated that ALC (0.01 mM) was

able to significantly prevent GLUT3 decrease, probably by donating an acetyl group to

glucosamine, promoting its stabilization. Importantly, in the present study, increased

glucose uptake in PC12 cells exposed to ALC and METH, resulted in decreased cell

viability, which was associated with early (24h) reduction of ATP synthesis, but seems to

protect mitochondrial function later on (72h). Conversely, in a high-glucose medium, ATP

synthesis in cells exposed to equal concentrations of ALC and METH was much higher.

These results suggest that when glucose is available, ALC promotes glycolysis for energy

production, but if glucose is scarce, ALC may promote alternative metabolic pathways,

such as β-oxidation of fatty acids (Flanagan et al., 2010; Abdul Muneer et al., 2011b). In a

glucose-deprived conditions, METH seems no to affect ATP production, that was seen in

primary neuronal cells, where no clear evidence of METH action over ATP synthesis was

observed (Abdul Muneer et al., 2011b).

The binding of IGF1 to its receptor can act via different pathways, including the PI3-K–Akt,

which inhibits pro-apoptotic proteins of the Bcl-2 family and induces anti-apoptotic proteins

of the same family, connecting IGF1 and cell survival (Bondy and Cheng, 2004;

Fernandez and Torres-Aleman, 2012). Bax is a pro-apoptotic homologue of Bcl-2 protein

present in the cytosol, which is activated in apoptosis, undergoing conformational

modifications and translocation to the outer mitochondrial membrane or to the nucleus

(Dewson et al., 2001). Here, although the ratio of activated Bax was unaltered, ALC alone

or in combination with METH, led to the translocation of active Bax mostly to the nucleus.

Although Bax may be associated with the nuclear matrix even in the absence of an

Page 94: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

94

apoptotic stimulus (Wang et al., 1999; Motyl et al., 2000), Bax translocation is commonly

seen as an early event of apoptosis (Gajkowska et al., 2001; Gill et al., 2008). Adding to

this, we observed that Bax translocation was concomitant with decreased ∆ψm and higher

ROS formation. ROS are important mediators in mitochondrial apoptotic signaling (Cande

et al., 2002). ALC is often seen as an energy enhancer, improving cellular oxygen

consumption, which can increase oxidative stress. In a number of pathologic conditions,

electron leakage augmenting ROS production is preceded by bioenergetic impairment due

to loss of mitochondrial function (Adams et al., 2001; Virmani et al., 2004).

In summary, the present results show that ALC preconditioning in mice exposed to a low

METH dose led to a robust increase in glucose uptake that was concomitant with impaired

reference memory in the WM. Furthermore, in vitro, ALC/METH combination led to

decreased viability, reduced ATP synthesis, increased ROS and loss of mitochondrial

membrane potential. The translocation of active Bax to the nucleus can be associated

with apoptotic events.

Page 95: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

CHAPTER 3

NEUROPROTECTIVE ACTION OF ALC IN

DOPAMINERGIC NEUROTRANSMISSION

Page 96: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 97: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

97

3.1. INTRODUCTION

As stated before, pre-treatment with ALC revealed a neuroprotective effect against

MDMA-induced neurotoxicity, not only at the mitochondrial level, but it also prevented a

MDMA-induced decrease of serotonin levels in several regions of the rat brain (Alves et

al., 2009). However, the molecular mechanisms underlying the mechanism of action of

ALC, particularly regarding the induction of changes in neurotransmitter systems are still

not fully understood. This section aims to contribute to a better understanding of these

mechanisms and explore the potential neuroprotective effect of ALC in the therapy of

neurodegenerative disorders that result from mitochondrial dysfunction.

Exposure to METH provides an excellent model to mimic these neurodegenerative

processes, both in vivo and in vitro (Ferrucci et al., 2008a). Using a cell line model of

exposure to METH, we aim to contribute to clarify the mechanism by which administration

of ALC alters neurotransmitter release. METH exposure is well known to induce altered

function of the dopaminergic system, initially increasing striatal DA content, which

increases oxidative stress, leading to intracellular alterations at the nigral DA cell bodies,

followed by degeneration of dopaminergic terminals and decreased striatal release of DA

(Ferrucci et al., 2008b). These features of METH action provide an excellent model to

study the action of ALC over DA release.

As mentioned in the first chapter, the PC12 cell line derived from the rat

pheochromocytoma is well known for its catecholamine production and is often used as

an in vitro model to mimic dopaminergic function and may predict the neurotoxicity of a

variety of compounds acting on central dopaminergic neurons (Schlegel et al., 2004; Piga

et al., 2005). In the present study, aiming to evaluate the putative role of ALC over the

function of neurotransmitter systems, we have exposed PC12 cells pre-treated with ALC,

to different concentrations of METH, for either 24h or 72h. The intra- and extracellular

levels of DA were assessed to evaluate the influence of ALC on dopaminergic function.

A differential effect of METH and DA itself on post-synaptic neurons was previously

demonstrated (Fornai et al., 2007). DA receptors seem to have a pivotal role in DA-

induced cell death. As D2 receptors (D2R) are well known to function as auto-receptors

and are also possible modulators of D1 receptors, we aim to identify the putative

protective role of ALC in D2R mediated METH neurotoxicity. Since the D2R play an

important role in the release of DA, a potential effect of ALC on the function of these

receptors may be relevant to improve our understanding of the mechanisms of action of

ALC as a potential neuroprotective drug for dopaminergic neurons. In the present work we

Page 98: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

98

also aim to verify the presumed role of D2R availability as a mechanism of action of ALC’s

ability to protect dopaminergic toxicity through DA receptor imaging with the radiotracer

iodine-123-labeled (S)-2-hydroxy-6-methoxy-N-((1-ethyl-2-pyrrolidinyl)methyl) benzamide

(123I-IBZM). This radiotracer was first proposed by Kung et al. for the assessment of D2R

availability (Kung et al., 1988), exhibiting a high specificity to these receptors with no or

minimal metabolism (Kung et al., 1989). 123I-IBZM is a DA D2/3R antagonist exhibiting

high selectivity and moderate affinity to these receptors (KD D2R = 0.43 nM) (Kung et al.,

1989), making it a suitable agent to image DA release in regions of high D2R density

(Laruelle et al., 1995).

3.2. MATERIAL AND METHODS

3.2.1. Cell Culture and Drug Regimen

Rat pheochromocytoma PC12 cells (American Type Cell Culture Collection, Manassas,

VA) were used as an in vitro model to study neuroprotective effects of ALC. The cells

were used in their undifferentiated form between passage 12 and 16. Cells were grown in

Dulbecco’s Modified Eagle Medium (DMEM + GlutaMAX™-I, Gibco®) supplemented with

10% of fetal bovine serum (FBS, Gibco®) and 1% of penicillin (50 IU/mL) streptomycin (50

mg/mL) (PS, Gibco®) in a CO2 incubator at 37°C, with 5% CO 2 and 95% filtered air. Cells

in log phase were treated with increasing doses of ALC (0.01, 0.1, 0.5 and 1.0 mM) alone

or in combination with METH 1.0 or 100 µM. When ALC and METH were combined, pre-

treatment with ALC preceded METH exposure in 30 min. The selected doses of METH

were previously described and well characterized (Fornai et al., 2004; Fornai et al., 2007).

ALC was kindly provided by Sigma-Tau, SpA (Pomezia, Italy). METH hydrochloride was

obtained from Sigma-Aldrich® (St. Louis, MO). All subsequent experiments were

performed using culture medium without FBS in order to counteract the protective effect

conferred by the serum.

3.2.2. Dopamine Quantification

Cells were grown in 24-well plates (100,000 cells/well) and incubated with increasing

doses of ALC (0.01 mM to 1.0 mM), alone or in combination with METH 1.0 µM or 100

µM. Drugs were dissolved directly into the culture medium and incubated for 24h or 72h.

Cell culture medium was collected and stored at -80º C and cells were trypsinized and

Page 99: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

99

centrifuged. The pellets were re-suspended in perchloric acid (HClO4) 0.1 N, disrupted by

ultrasonication and centrifuged at 15,000 g for 5 min. Supernatant was collected and

filtered through a 0.2 µm nylon filter (Costar micro-centrifuge filter, Corning, NY). When

cell culture medium was defrosted, an equal volume of HClO4 0.2 N was added, disrupted

by ultrasonication and centrifuged at 15,000 g for 5 min. The supernatant was collected

and filtered through a 0.2 µm nylon filter (Costar micro-centrifuge filter, Corning, NY). DA

content was determined by high performance liquid chromatography, combined with

electrochemical detection (HPLC-EC) as previously described (Alves et al., 2009).

Aliquots of 50 µl were used to assay DA levels by HPLC-EC using a Gilson instrument

(Gilson, Inc., Middleton, WI, USA), fitted with an analytical column (Supelco Supelcosil

LC-18 3 µM; 7.5 cm x 4.6 mm; flow rate: 0.8-1.0 mL/min; Supelco, Bellefonte, PA, USA).

A mobile phase of 0.7 M aqueous potassium phosphate (monobasic) (pH 3.0) in 10%

methanol, 1-heptanesulfonic acid (222 mg/L) and Na-EDTA (40 mg/L) was used.

Concentrations of DA were calculated using a standard curve. Dopamine standard was

purchased from Sigma (St. Louis, MO, USA). Final results were expressed in terms of

monoamine content per cell to allow direct comparison between intra- and extracellular

levels.

3.2.3. In Vivo Quantification of Dopamine Striatal Binding

Twelve week-old male C57BL/6J mice (21.6 – 26.0 g) were purchased from Charles River

(Innovo, Gödöllı, Hungary) and maintained on a 12-h light/dark cycle. All experimental

procedures were approved by the Local Animal Experimental Ethical Committee of

Semmelweis University, Budapest, Hungary, in compliance with the European Community

Council Directive of September 22, 2010 (2010/63/UE). All efforts were made to ensure

minimal animal stress and discomfort. Room temperature was kept at 22ºC and animals

were allowed water and food ad libitum. Twelve animals were divided into four groups,

according to the different treatments: group 1 (control, saline), group 2 (ALC, 100 mg/kg),

group 3 (METH, 10 mg/kg) and group 4 (ALC + METH 30 min later). Drugs were

administered by intraperitoneal injection. Thirty minutes after the last dose, 14.2 ± 0.9

MBq 123I-IBZM (General Electric Healthcare, Eindhoven, the Netherlands) with a specific

activity higher than 74 TBq/mmol (radiochemical purity >95% and radionuclide purity

>99.9% at the reference time) was injected in the tail vein, under anesthesia (isoflurane

2%). After injection, animals recovered very quickly from isoflurane anesthesia and were

returned to their home cage for the radiotracer uptake period (70 minutes). It was

previously shown that specific striatal 123I-IBZM binding in the human and rat brain, is

Page 100: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

100

maximal at approximately at 40 minutes after injection, remaining stable for up to 2 hours

(Verhoeff et al., 1991b; Verhoeff et al., 1991a). In this study, image acquisition started 70

minutes after radiotracer intravenous injection and prolonged for 45 minutes. Three

consecutive SPECT acquisitions were performed at 70, 85 and 100 minutes after

radiopharmaceutical intravenous injection. Immediately before image acquisition, animals

were anaesthetized with isoflurane 2%, the head was fixed to a specific head holder bed

(MultiCellTM, Mediso, Hungary) and the body immobilized with tapes to prevent movement.

Images were acquired in a NanoSPECT/CTTM system (Mediso/Bioscan, Budapest,

Hungary). Multiplexing multi-pinholes were used (9 in total; 0.7 mm diameter apertures)

were used (SciVis, Göttingen, Germany). Thirty circular SPECT projections over a full

circle were collected each for 84 seconds in a 159 keV ± 20% photopeak energy window.

Projections were reconstructed into three-dimensional images using the HiSpect software

(SciVis, Göttingen, Germany) with a proprietary 3D OSEM algorithm, in 0.2 x 0.2 x 0.2

mm voxels with decay correction. Magnetic resonance images (MRI) were acquired in a

PET-MRI scanner (nanoScan PM, Mediso, Hungary), equipped with one Tesla magnet for

25 minutes. All images were co-registered and visualized using Interview FusionTM

(Mediso, Hungary) and VivoQuant (inviCRO, Boston, USA) dedicated image processing

software. We also used advanced post processing Hybrid Viewer™ software, courtesy

from HERMES Medical Solutions (Stockholm, Sweden). Regions of interest (ROI) were

drawn over the left and right striatum as well as in the cerebellum in order to determine

the striatal binding ratio (expressed as the quotient between striatal binding minus

cerebellar binding divided by cerebellar binding) (Jongen et al., 2008). For 123I-IBZM

uptake quantification, the secular equilibrium method was used, since there is no need of

plasma sampling nor the application of a reference region based on dynamic image

acquisition. Since the chosen radiotracer (123I-IBZM) presents an adequate time period of

binding equilibrium, it is possible to inject a single bolus instead of a continuous infusion

(Scherfler et al., 2005).

3.2.4. Statistical Analysis

A one-way analysis of variance (ANOVA, treatment) was used to analyze differences

between treatments. Significant differences were further tested using the post-hoc Tukey

honest significant differences (HSD). Differences were considered to be statistically

significant at p<0.05 level. All analyses were performed using Prism 6 for Mac OX

(GraphPad Software, San Diego, California, USA).

Page 101: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

101

3.3. RESULTS

3.3.1. ALC Action on Dopamine Release in PC12 Cells

After 24h of incubation, as described above, intracellular levels of DA were shown to be

significantly increased in cells treated with ALC 0.5 mM (one-way ANOVA, F(4,12)=6.152,

p=0.085, Tukey’s post-hoc). At this time point, no other differences compared to the

control condition were observed, either in intra- or extracellular levels of DA (figure 3.1). In

contrast, at 72h, the levels of intracellular DA were increased in PC12 cells treated with

ALC 0.01 and 0.1 mM (one-way ANOVA, F(4,17)=43.950, p<0.0001, for both conditions,

Tukey’s post-hoc) and still, no changes were observed in extracellular DA amounts (figure

3.1). This seems to indicate that ALC itself is able to increase the production of DA in

PC12 cells, without interfering with DA release and uptake processes.

Figure 3.1 – Effects of pre-treatment with ALC at 0 .01, 0.1, 0.5, 1.0 mM on DA release .

Graphic representation of intra- and extracellular levels of DA measured by HPLC-EC, in PC12

cells, when treated with ALC for 24h or 72h. Columns represent mean+SEM, expressed as pg of

neurotransmitter/cell for each experimental condition (n=6-9). Significant differences are marked as

**p<0.01 and ***p<0.001, as compared with the control condition (determined by one-way ANOVA

followed by a post hoc Tukey’s HSD).

Page 102: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

102

3.3.2. ALC Neuroprotective Action over METH 1.0 µM Exposure

Exposure to METH 1.0 µM is known to induce, within a few hours, the formation of

intracellular multilamellar inclusions that are both α-synuclein and ubiquitin-positive,

without a substantial impact on DA function (Ferrucci et al., 2008b; Fornai et al., 2008).

Accordingly, in the present work, a 24h exposure to METH 1.0 µM did not induce any

significant alteration in the intra- and extracellular levels of DA (figure 3.2).

However, when cells were exposed to METH 1.0 µM for a longer period (72h), a

significant increase in the intracellular levels of DA was apparent (one-way ANOVA,

F(5,257)=10.490, p<0.0001, Tukey’s post-hoc). Interestingly, when cells were pre-treated

with ALC, before exposure to METH, this increase was reduced and only reached

significance in cells pre-treated with ALC 0.5 mM (p<0.0001) (figure 3.2). At 72 hours,

extracellular levels of DA in cells exposed to METH 1.0 µM were increased when

compared to control conditions (one-way ANOVA, F(5,25)=4.751, p=0.0256, Tukey’s post-

hoc). Importantly, ALC 0.01 mM was able to decrease DA extracellular levels induced by

METH (p=0.0305). Furthermore, in cells pre-treated with other ALC concentrations, the

extracellular levels of DA almost reached significance when cells were treated with the

highest dose (ALC 1.0 mM, p=0.0771, figure 3.2). It is noteworthy that at this time point,

the control levels of extracellular DA were markedly increased when compared with the

intracellular DA levels (t test, p<0.0001). This reflects an increased release of DA through

the incubation period.

Page 103: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

103

Figure 3.2 – Effects of pre-treatment with ALC at 0 .01, 0.1, 0.5, 1.0 mM and METH 1.0 µM on

DA release . Graphic representation of intra- and extracellular levels of DA measured by HPLC-EC,

in PC12 cells pre-treated with ALC and exposed to METH 1.0 µM for 24h or 72h. Columns

represent mean+SEM, expressed as pg of neurotransmitter/cell for each experimental condition

(n=6-9 in control and METH1 conditions, and n= 3 replicated experiments for combinations of

METH 1 and ALC). Significant differences are marked as follows: *p<0.05 and ***p<0.001 as

compared with the control condition; and •p<0.05 as compared with METH 1.0 µM (determined by

one-way ANOVA followed by a post hoc Tukey’s HSD).

3.3.3. ALC Neuroprotective Action over METH 100 µM Exposure

Exposure to METH 100 µM is known to cause substantial cell loss, paralleled by massive

DA release (Fornai et al., 2004). In the present work, when cells were exposed to METH

100 µM, the expected decrease in intracellular DA levels was clearly observed (one-way

ANOVA, F(5,18)=182.4, p=0.0034, Tukey’s post-hoc). Notably, all tested doses of ALC

were effective in preventing the METH-induced decrease of DA concentrations

(p<0.0001). Moreover, in cells pre-treated with ALC 0.1 mM, 0.5 mM and 1.0 mM,

intracellular levels of DA at 24h was markedly increased when compared to the control

p=0.0771

Page 104: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

104

(p<0.0001, figure 3.3). However, at this time point, the levels of DA in the extracellular

medium were unaltered for all tested conditions. Consequently, these results indicate that

ALC not only effectively prevented the METH-induced decrease of DA concentrations, but

also increased DA production.

At 72h, exposure to METH 100 µM led to increased intracellular levels of DA (one-way

ANOVA, F(5,26)=5.046, p=0.0192, Tukey’s post-hoc), which were prevented by all tested

doses of ALC except 0.1 mM (figure 3.3). As observed for METH 1.0 µM, a 72h exposure

caused a massive release of DA into the extracellular medium (one-way ANOVA,

F(5,24)=4.961, p=0.0113, Tukey’s post-hoc). In spite of this, in cells pre-treated with ALC,

extracellular levels of DA were not increased when compared to the control, except for the

lowest dose of ALC. Interestingly, this same ALC dose was the most effective in

preventing DA loss after exposure to METH 1.0 µM.

Figure 3.3 – Effects of pre-treatment with ALC at 0 .01, 0.1, 0.5, 1.0 mM and METH 100 µM on

DA release . Graphic representation of intra- and extracellular levels of DA measured by HPLC-

EC, in PC12 cells pre-treated with ALC and exposed to METH 100 µM for 24h or 72h. Columns

represent mean+SEM, expressed as pg of neurotransmitter/cell for each experimental condition

Page 105: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

105

(n=6-9 in control and METH100, n= 3 replicated experiments for combinations of METH100 and

ALC). Significant differences are marked as follows: *p<0.05; **p<0.01 and ***p<0.001 as

compared with the control condition; and •••p<0.001 as compared with METH (determined by one-

way ANOVA followed by a post hoc Tukey’s HSD).

3.3.4. ALC Neuroprotective Action over DA Receptors In Vivo

Drug-occupancy studies are often performed to evaluate the selectivity of drugs to specific

brain regions, using radiopharmaceuticals with high specific activity in tracer amounts

(such that radiotracer occupancy represents less than 5% of the total available receptor

sites) (Hume et al., 1998; Henriksen and Drzezga, 2011). In the present study, 123I-IBZM

was used to evaluate D2R occupancy in C57BL/6J male mice, after the administration of

ALC alone or in combination with METH, a DA agonist. Images were acquired in three

different time points with 15 minutes intervals, starting 70 minutes after 123I-IBZM injection.

A single dose of METH induced a significant decrease of striatal 123I-IBZM binding in both

the left and right striatum that was evident since the first acquisition point (i.e., 70 min

post-injection) [two-way ANOVA (treatment x acquisition time), F(6,24)=0.7784, p<0.0001

for both factors, but no interaction between them].

In the left striatum, there was a progressive displacement of the radiotracer binding over

time (two-way ANOVA followed by a post-hoc Tukey’s test F(3,24)=21.07, p=0.0336

comparing to control at T70 and p=0.0012 at T100, figure 3.4-A). Interestingly, over time,

ALC was able to reverse the decrease on the radiotracer binding induced by METH. At

the beginning of image acquisition period, the ALC/METH group presented a significant

decrease in radiotracer binding ratios (p=0.0024 to control and p=0.0116 to ALC, figure

3.4-A), but in the end, this condition was no longer different from control.

The right striatum was less affected than the left, but the progressive decreasing pattern

induced by METH was similar to that observed in the left striatum. From the 85th minute

after tracer injection and onwards, 123I-IBZM binding in the right striatum was significantly

reduced comparing to the control condition (two-way ANOVA followed by a post-hoc

Tukey’s test F(3,33)=13.26, p=0.0221 at T85 and p=0.0012 at T100, figure 3.4-B). In this

case, although less evident, ALC was also able to reverse the decrease in 123I-IBZM

binding induced by METH. However, conversely to the left striatum, at 85 minutes after

radiotracer administration, in the right striatum ALC still did not reversed this decrease,

which was only visible 100 minutes after tracer injection (figure 3.4-B). ALC per se did not

Page 106: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

106

interfere with D2R binding, since the average binding ratio was always similar to the

control group (figure 3.4-A and B).

Figure 3.4 – ALC reverses the METH-induced increase d D2 receptors occupancy . A: Striatal

binding ratio of 123I-IBZM in the left striatum. B: Striatal binding ratio of 123I-IBZM in the right

striatum. It is evident the significant progressive radiotracer displacement in the D2R induced by

METH in both striatal regions. Right striatum was less affected than the left. ALC was able to

reverse the radiotracer displacement induced by METH. Significant differences are marked as

*p<0.05 and **p<0.01, as compared with the control condition; �p<0.05 and ��p<0.01 as

compared with ALC (as determined by one-way ANOVA, followed by a post-hoc Tukey’s test).

In figure 3.5 representative tomographic coronal slices from SPECT image acquisition of

mice brain are shown. Images clearly demonstrate a sharp decrease of striatal 123I-IBZM

binding in the METH group comparing to all other groups.

Page 107: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

107

Figure 3.5 – Representative 123I-IBZM SPECT-MRI images from 12-week old male mice . The

displayed tomographic images were acquired 100 minutes after radiotracer injection and were

reconstructed by OSEM algorithm. The coronal planes, representative of each group are shown.

Note the decrease on radiotracer uptake on METH group, which is increased when animals were

preconditioned with ALC.

3.4. DISCUSSION

The present study uses both a cell line and an animal model of neurotoxicity to evaluate

the action of ALC on the dopaminergic neurotransmission system. It revealed that, in vitro,

ALC can interfere with DA release, decreasing the amount of extracellular DA in the

presence of METH. Extracellular DA may be neurotoxic to dopaminergic neurons. These

data substantiate those obtained in vivo in C57BL/6J mice in which ALC was able to

counteract the effects of METH on D2R occupancy by increasing the receptor

displacement of METH. In fact, METH is well-known to increase the synaptic

concentration of DA (Di Chiara and Imperato, 1988), resulting in an increased receptor

occupancy (DA competes with the radiotracer for D2R binding) and thus, reduces the

availability of receptors for radiotracer binding. There is extensive imaging evidence that

amphetamine, by promoting DA release, reduces the in vivo binding of imaging

radiotracers that are either D2R agonists (3H-N-propylnorapomorphine) (Kohler et al.,

1981) or antagonists (123I-IBZM, 11C-raclopride) (Carson et al., 1997; Jongen et al., 2008),

but the counteracting effect of ALC was never reported.

Page 108: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

108

Using a rat model of MDMA exposure, our group observed that pre-treatment with a single

dose of ALC (100 mg/kg) was effective in preventing a marked serotonin loss in all

assessed brain regions, indicating a clear ALC protective effect (Alves et al., 2009).

Although ALC treatment seems not to affect healthy subjects, where neurotransmitter

levels are kept mostly within the normal ranges (Adriani et al., 2004), in regions like the

hippocampus or the ventral mesencephalon, our group observed increased levels of

serotonin in rats treated only with ALC (Alves et al., 2009). The physiologic significance of

this remains unclear.

In the present study, when cells were treated with a range of ALC doses, but not exposed

to METH, intermediate doses of ALC led to increased levels of intracellular DA.

Interestingly, this was observed either at 24h or 72h, although the degree of response

varied with different doses of ALC. Moreover, at both time points, the extracellular levels

of DA were unaltered throughout tested conditions, which support the idea that ALC

treatment, in the absence of a neurotoxic event, may lead to increased synthesis of DA.

ALC has long been reported to affect the cholinergic, serotonergic, dopaminergic,

glutamatergic and GABAergic systems, but the mechanisms involved were mostly

unknown (Tolu et al., 2002). Recently, Scafidi et al. using 13C-ALC, demonstrated that the

acetyl moiety of ALC is used as an energy source by both astrocytes and neurons and

rapidly incorporated into glutamate and GABA synthesis (Scafidi et al., 2010b). This

clarified the direct involvement of ALC in increased glutamine, glutamate and GABA

levels, via the TCA cycle. Importantly, the biosynthesis of both tyrosine and L-tryptophan,

as well as DA and 5-HT precursors, involves either a transamination reaction with

glutamate or the participation of glutamine as an amine donor (Yudkoff, 1997), which may

explain the action of ALC over monoamine production. Noteworthy, glutamate is also the

precursor of glutathione biosynthesis (Griffith and Mulcahy, 1999), an enzyme implicated

in the antioxidant properties of ALC, as explained in chapter 1.

The action of METH over PC12 cells was extensively characterized. It was demonstrated

that increased endogenous DA is required for METH-induced formation of ubiquitin and α-

synuclein positive lamellar bodies (Fornai et al., 2004). These inclusions are not

necessarily associated with neurodegeneration and are thought to represent a defense

mechanism, against higher levels of oxidized and misfolded proteins. This seems to be

confirmed in PC12 cells, where exposure to 1.0 µM of METH dose, produces the highest

number of inclusions without cell death, while after exposure to 100 µM METH, inclusions

are not visualized and massive cell damage occurs (Fornai et al., 2004). Interestingly, DA

synthesis may promote inclusion formation either directly or via alternative metabolic

Page 109: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

109

pathways, since production of these inclusions can be avoided both by DA depletion or

other processes, such as administration of the iron chelator, antioxidant agent S-

apomorphine (Fornai et al., 2004). In this work, in accordance with previous reports, a 24h

exposure to METH 1.0 µM did not affect DA production and release. However, at 72h,

both intra- and extracellular DA levels were increased. Pre-treatment with ALC (except for

the 0.5 mM dose) led to intracellular DA levels that were no longer significantly higher

than in the control condition. Moreover, while METH exposure induced a marked release

of DA, pre-treatment with ALC seems to interfere with DA release, decreasing the amount

of extracellular DA. These results were obtained for in vitro assays with PC12 cells as well

as for in vivo with C57BL/6J male mice, in which a single dose of METH (10 mg/kg) led to

a significant increase in DA release into the synaptic cleft, which then competes with 123I-

IBZM for binding to D2R. In striatal neurons, increased extracellular or extravesicular DA

levels were shown to occur in concomitance with increased levels of ROS (Cadet and

Brannock, 1998; Lazzeri et al., 2007). In the brain, catecholamines are indeed an

important source of free radicals as its autoxidation induces the production of quinones

and, subsequently, the generation of free radicals and damage to proteins (Cadet and

Brannock, 1998).

Administration of METH 1.0 µM to the cell culture medium corresponds to an in vivo dose

of 5 mg/kg, which leads to a striatal concentration of about 1.0 µM (Fornai et al., 2007). As

already mentioned in the first chapter, DA release in PC12 cells is much lower than in

striatal cells due to poorer storage and synthesis. Furthermore, while increasing METH

dose in vivo leads to loss of striatal terminals but not to soma loss, increasing METH dose

in PC12 cells induces both necrosis and apoptosis (Fornai et al., 2007), which renders it

difficult to directly compare in vivo exposure with PC12 cells exposure to METH. On the

other hand, PC12 cells, like DA neurons, present the type A isoform of MAO, providing a

good model for the study of altered redox events related to DA release. Therefore,

increasing exposure from 24h to 72h, in PC12 cells, mimics the increase in ROS

formation induced in vivo by METH exposure and provides an interesting model for the

study of ALC properties.

In this study, the challenge with a single dose of 10 mg/kg of METH decreased striatal

D2R binding ratios relative to the control group between 20% and 30%, depending on the

time point of image acquisition. Similar results were obtained by Jongen et al., who

observed a reduction in striatal D2R binding ratios of 27% in mice after the administration

of 2.5 mg/kg of amphetamine (Jongen et al., 2008). In another study, in which

amphetamine and the dopamine reuptake inhibitor methylphenidate were compared,

Page 110: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

110

microdialysis assays showed that amphetamine (2.5 mg/kg) induced a 1400% increase in

extracellular DA, while methylphenidate (5 mg/kg) increased it in 360%. This four-fold

difference was not paralleled by synaptic DA levels, because reductions of striatal D2R

binding after the administration of amphetamine and methylphenidate were 24 and 21%,

respectively, as measured by 11C-raclopride (an antagonist D2R radiotracer) displacement

(Schiffer et al., 2006). This may be due to the distinct pharmacologic mechanisms

underlying these drugs actions. While methylphenidate action is confined to the synaptic

cleft, by blocking the passive reuptake of DA, amphetamine binds to DAT, thus entering

the presynaptic terminal and actively releasing high amounts of vesicular DA, inducing a

massive increase in the ratio of extracellular to synaptic DA, comparing to

methylphenidate (Schiffer et al., 2006). In human studies, METH was able to reduce D2R

availability in the caudate (16%) and in the putamen (11%), similarly to observations for

cocaine abusers (Volkow et al., 2001d). In schizophrenic patients with schizotypal

personality disorder, METH also induced a significant decrease (12%) in 123I-IBZM striatal

binding ratios, which was more pronounced during illness exacerbation (24%) (Abi-

Dargham et al., 2004). Another interesting aspect of METH action is related with altered

membrane permeability. Recent studies have confirmed that METH inhibits potassium-

dependent DA release, likely caused by diminished vesicular DA uptake (Chu et al.,

2010). Conversely, increased membrane permeability was long implicated in the acute

effects of METH and massive leakage of albumin was shown to occur across the BBB in

the cerebral cortex of METH-treated mice, concomitantly with nerve cell damage (Sharma

and Ali, 2006; Martins et al., 2011). Whether decreased radiotracer binding to D2R after

METH administration is due to competitive inhibition with DA or to D2R internalization

(Sun et al., 2003; Skinbjerg et al., 2010) or even to changes in receptor affinity (Ginovart,

2005) is a matter of controversy, although the competitive inhibition theory has been the

most consensual (Jongen et al., 2008). The intracellular milieu has lower pH and sodium

ion concentration comparing to the extracellular environment. In these conditions, the

affinity of benzamide-derived D2R radiotracers is lower, explaining the lower radiolabelled

benzamides uptake when the receptors are internalized due to the action of DA (Shotbolt

et al., 2012). Nevertheless, decreases on radiotracer binding to D2R relative to baseline

provide reliable estimations concerning increases on synaptic DA concentration (Nikolaus

et al., 2005).

In another study with baboons, pre-treatment with vigabatrin restored to normal levels the

striatal 11C-raclopride (D2R antagonist) binding after acute cocaine administration (Dewey

et al., 1997). Vigabatrin, similarly to ALC in the present study, was able to decrease the

extracellular DA concentrations induced by cocaine (Dewey et al., 1997). The

Page 111: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

111

administration of ALC to rats for seven consecutive days shown to increase DA in the

nucleus accumbens shell which was associated to protective effect on acute stress

exposure (Tolu et al., 2002).

A critical issue in brain receptor imaging is related to radiotracer specific activity (i.e., the

radioactivity per mass unit). In order to assure that receptor occupancy is less than 5%, a

high specific activity should be assured (Hume et al., 1998), otherwise significant receptor

occupancy or eventually, pharmacological effects might occur, invalidating study results

(Franc et al., 2008). In the present work, a radiopharmaceutical with a high specific activity

(> 74 TBq/mmol) was used in order to avoid not only the occupancy of a great number of

D2 receptors, but also pharmacological effects to occur. Additionally, the use of a high

sensitivity micro-SPECT camera allowed the administration of rather low doses of 123I-

IBZM.

A practical requirement related to in vivo animal imaging is that the animal has to remain

still during the imaging procedure, requiring most often the administration of anesthetizing

agents. However, it is reported that binding of radioactive tracers to brain target regions

may be affected by anesthetics. Isoflurane was shown to induce changes in the

dopaminergic system in nonhuman primates, possibly by modulating the availability of the

dopamine transporter (Kobayashi et al., 1995; Tsukada et al., 1999). Furthermore,

halothane anesthesia enhanced METH-induced DA release in the rat brain (Adachi et al.,

2001). However, other studies exclude a significant effect of isoflurane anesthesia on D2R

imaging (signal to noise ratio and radiotracer uptake) (Honer et al., 2004; Schiffer et al.,

2006). Our protocol was based on a short inhalation period of a light dose of isoflurane

(2%), which started immediately before the intravenous administration of the radiotracer

and lasted only the time required for the injection procedure. After injection, animals

recovered very quickly from isoflurane anesthesia and were returned to their home cage

for the 123I-IBZM uptake period (70 minutes). Animals were anesthetized again for the

imaging acquisition procedure. Since during the uptake period, animals were no longer

under the effect of the anesthetic, we might assume that anesthesia did not have a

significant effect on DA release.

The administration of repeated high doses of METH and amphetamine has been shown to

not only affect receptors availability, but also to cause long lasting DAT depletion (Wagner

et al., 1980). However, amphetamine seems to be more effective than METH in

decreasing the number of DAT (Booij et al., 2006). Volkow et al. observed a significant

DAT reduction in human METH abusers that persisted at least for 11 months after

detoxification. Moreover, in these subjects, DAT reduction was associated with motor and

Page 112: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

112

cognitive impairment (Volkow et al., 2001c). DAT are crucial for the termination of

dopaminergic transmission through the reuptake of DA from the synaptic cleft and thus,

have an important role in the spatial and temporal regulation of synaptic DA (Booij and

Kemp, 2008). In the recent years, some authors came up with the idea that

amphetamines might promote the fast internalization of DATs with the consequent

reduction on the surface expression to bind to the DAT radiotracer (ex: 123I-ioflupane)

(Saunders et al., 2000). In this study, we have used a single and relatively low dose of

METH, which might be insufficient to induce such significant changes on DAT.

A limitation of this study is related with the low number of imaged animals per group (n=3).

In order to increase the statistical significance of the study, and thus the respective power,

the number of animals per group should have been, ideally, at least 5. Moreover, in order

to validate these in vivo imaging results, it would be worth to use another imaging

technique, the ex-vivo autoradiography (this is an ongoing task, since we have already

started it). This would allow the comparison between the striatal binding ratios obtained by

both imaging techniques. Also, the evaluation of DA levels in the synaptic cleft by

microdialysis technique would bring valuable information concerning the competitive

inhibition and the receptors internalization theories. Due to the possible effects of ALC on

D1 receptors (D1R) and the influence of D2R on these receptors, the evaluation of those

receptors could be interesting. However, so far, the availability of radioactive tracers to

assess D1R is very limited. Furthermore, the evaluation of intra- and extracellular DA

levels (and its metabolites, such as 3,4-dihydroxyphenylacetic acid, DOPAC, and

homovanillic acid, HVA) in primary cultures of neurons would allow to complement and

validate data obtained in PC12 cells.

3.5. CONCLUSION

The present study demonstrates a possible effect of ALC over METH-induced DA release.

Our results show that ALC by itself increases intracellular levels of DA, which might be a

consequence of the ALC role in tyrosine biosynthesis. However, in vivo, pretreatment with

ALC was shown to decrease D2R occupancy subsequent to METH administration. The

exact mechanisms underlying the action of ALC over DA release and membrane transport

remain unclear. However, it is possible that ALC may exert its protective role through the

direct regulation of cytoskeleton properties, which may interfere with the conformation of

membrane receptors and therefore affect its binding affinity to different compounds.

Alternatively, ALC may induce changes in the expression of DA receptors, and particularly

Page 113: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

113

the D2R, since this receptor is crucial for the regulation of DA release, and consequently

may play an important role in the induction of dopaminergic toxicity.

Page 114: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 115: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

CHAPTER 4

GENERAL DISCUSSION

Page 116: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 117: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

117

In order to better understand the mechanisms underlying the action of ALC, we have

focused our work on two main aspects: a) the role of ALC in mitochondrial functionality in

vitro and glucose uptake, both in vitro and in vivo; b) the role of ALC over the

dopaminergic function, both in vitro and in vivo.

Carnitine has been described has having a key role in cellular bioenergetics not only by

facilitating the transport of fatty acids across the mitochondrial membrane into the matrix,

where they undergo β-oxidation, but also by controlling the intramitochondrial ratio of

acetyl-CoA/free CoA and consequently, modulating glucose oxidation. Considering these

roles in cellular bioenergetics, allied to the reported putative neuroprotective action, we

proposed to study the mechanisms involved in ALC action, particularly those related to

mitochondrial functionality and dopaminergic function. Since the brain relies in glucose

oxidation as the main source of energy, we started by assessing the role of ALC in

neuronal metabolism in the mice brain in vivo, under several experimental conditions. We

used a radioactive analogue of the glucose molecule (18F-FDG) to image C57BL/6J young

male mice. On one hand, the expression of IGF1 seems to modulate glucose uptake

through the indirect promotion of GLUTs translocation to the plasma membrane,

augmenting glucose transport into the cell. On the other hand, carnitine supplementation

activates the IGF1 signaling cascade and, consequently, promotes glucose utilization.

These assumptions led us to investigate the expression of both Igf1 and Igf1R in the

striatum, where the most relevant changes in glucose uptake were observed. To

complement in vivo data, we used a dopaminergic cell line to better understand the role of

ALC in neuronal glucose uptake under the action of METH, in glucose-deprived

conditions. Regarding glucose uptake, our results showed:

o a generalized increase in glucose uptake in several brain regions, immediately

after the administration of METH when mice were preconditioned with ALC. This

was particularly evident in regions receiving a strong dopaminergic input and

therefore, more vulnerable to METH action (the striatum and the PFC);

o an increase in Igf1 levels in animals treated with ALC and a decrease in those

treated with METH;

o an upregulation of Igf1R levels in ALC/METH group, matching the increase in

glucose uptake and mismatching the Igf1 levels, that were unaltered;

o that the lowest tested dose of ALC (0.01 mM) increased glucose uptake, when

administered alone or in combination with METH.

According to our observations, the administration of either ALC or METH alone did not

show to have immediate (1-2 hours after injection) effects on glucose uptake. These

Page 118: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

118

findings were not surprising since the effect of an acute single low dose is not comparable

to a chronic exposure, which in the case of METH is known to induce significant

reductions in glucose metabolism in several brain regions. Conversely, the combined

administration of these drugs, increased glucose uptake in the mice brain, particularly high

in regions with important dopaminergic inputs. Although not addressed in the present

work, behavioral tests were held in the lab and animals that performed poorly in water

maze test, were those treated with ALC/METH, reflecting memory impairments.

Accordingly, Igf1R levels in the striatum of these animals were also increased 24h after

drug administration. However, the enhancement in Igf1R levels was not paralleled by

increases in Igf1 transcript levels. It is possibly that the systemic IGF1 may directly cross

the BBB and this may be the reason for unaltered Igf1 levels in the striatum. If this is the

case, IGF1 levels would also be increased. Increased Igf1 expression induced by ALC

may be an attempt to promote GLUTs translocation to the outer cell membrane, thus

augmenting glucose uptake by the brain, which seems to be relevant for the action of

ALC.

We reason that the combined action of ALC and METH over the dopaminergic and

adrenergic receptors and transporters, associated to neurotransmitters synthesis

promoted by ALC, may be in the basis of the observed increase in glucose uptake.

Moreover, our results indicate that the activation of IGF1 signaling cascade, namely the

increase in the IGF1R levels and the eventual indirect promotion of GLUTs translocation

to the plasma membrane, are also contributing factors.

To further explore the role of ALC in mitochondrial function, a dopaminergic cell line

(PC12 cells) exposed to METH in deprived glucose conditions (LG) was used. Here, we

assessed parameters such as cell viability, mitochondrial function and ATP synthesis. The

ability of ALC to protect cells from loss of mitochondrial membrane potential and prevent

the activation of the apoptotic cascade was also explored. Our results indicate that METH

significantly decreases mitochondrial function for a 72h exposure, which was partially

prevented when cells were preconditioned with ALC 0.01 mM. The combined use of this

dose of ALC and METH, which revealed a significant increase in glucose uptake at 24h, is

associated to decreased cell viability and ATP synthesis. On the other hand, when cells

were exposed to equal concentrations of ALC and METH in a high-glucose medium, ATP

synthesis was much higher. This suggests that if glucose is available, ALC promotes

glycolysis for energy production, but if it is scarce, it may promote alternative metabolic

pathways.

Page 119: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

119

Our findings are not clear concerning the activation of the apoptotic cascade, as we

observed no changes in the ratio of activated vs. total Bax. However, fluorescence images

showed that ALC/METH, led to the translocation of active Bax to the nucleus, paralleled

by decreased mitochondrial membrane potential (∆ψm) and increased ROS formation.

Our results suggest that increased ROS formation, loss mitochondrial membrane potential

and Bax translocation to the nucleus or mitochondria are likely to be early events in the

activation of the apoptotic cascade. Of note, we have also assessed the activation of

caspase 3 wihtout observing any significant change.

In order to accomplish our goals, we further evaluated the role of ALC over the

dopaminergic functionality, both in vitro and in vivo. The above mentioned dopaminergic

cell line and animal model were used and exposed to the same experimental conditions.

Intra- and extracellular DA levels were evaluated at 24h and 72h in PC12 cells. For animal

studies, an antagonist of D2R labeled with a radioactive isotope, was used to measure in

in vivo, the availability of these receptors. Our results reveal that, in vitro, ALC can

interfere with DA release, decreasing the amount of extracellular DA in the presence of

METH. These data corroborate with those obtained in mice in which ALC was able to

counteract the effects of METH on D2R occupancy by increasing the receptor

displacement of METH. ALC by itself increased intracellular levels of DA, which might be

a consequence of the ALC role in tyrosine biosynthesis. In vivo, pre-treatment with ALC

showed to decrease D2R occupancy by DA, released as a consequence of METH

administration. Possibly ALC exerts a protective effect through the direct regulation of

membrane channels or transporters or by inducing changes in the conformation or in the

expression of DA receptors, particularly the D2R, since they are crucial for the regulation

of DA release.

Our group previously reported that animals administered with ALC presented increased

levels of 5-HT (Alves et al., 2009) in the same brain regions that in the present study

displayed the highest glucose uptake. The administration of repeated high doses of METH

has been shown to not only affect receptors availability, but also to cause long lasting

DAT depletion. In this study, we have used a single and relatively low dose of METH,

which might be insufficient to induce such significant changes on DAT. In the future, it

would be interesting to study if ALC exerts some action over the DAT as it does over D2R.

To our knowledge, this aspect was not explored yet.

In vivo imaging requires that the animal does note move while is being scanned. This

usually involves the administration of anesthetizing agents. A matter of concern and

controversy is whether anesthetics modify radiotracers biodistribution and uptake level in

Page 120: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

120

the brain. There is evidence in the literature that those agents reduce 18F-FDG uptake in

several brain regions and may enhance the release of DA into the synaptic cleft. In the

present study we consider that isoflurane anesthesia might have had a minor or even no

effect in the radiotracers uptake due to the use of a low dose and a short period of

administration (limited to the time required for the intravenous injection procedure) of

isoflurane anesthesia, after which the animal recovered very promptly. During the

radiotracers uptake period (45 minutes for 18F-FDG and 70 minutes for 123I-IBZM) the

animals were no more under anesthesia.

A potential limitation of the present study is related with the low number of imaged

animals. In order to increase the power of the study, each group should have included at

least 5 animals. Despite this limitation, the obtained data presented a rather good

correlation with the remaining assays. Moreover, we think that the assessment of the

striatal levels of GLUTs and IGF1 would strengthen the data related to the Igf1, IGF1R

and glucose uptake. Regarding the dopaminergic function, we think that in the future, it

could be interesting to explore the role of ALC in D1R as well as in DAT (so far, these

aspects remain unexplored).

Overall, these data demonstrate that ALC exerts its action by increasing glucose uptake

and Igf1R levels in brain regions with strong dopaminergic input, such as the striatum after

an acute insult such as the exposure to METH. ALC by itself enhances the levels of Igf1.

Concerning neurotransmitter systems, ALC showed not only to increase the intracellular

levels of DA, but also to improve the availability of D2R by promoting METH displacement.

Although further studies are needed to support the use of ALC as a therapeutic agent,

based on our data and from others, we think that in the future, it might be possible to use

this compound as an adjuvant therapeutic agent in the treatment of some

neurodegenerative diseases.

Page 121: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

REFERENCES

Page 122: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential
Page 123: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

123

REFERENCES

Abdul Muneer PM, Alikunju S, Szlachetka AM, Haorah J (2011a) Inhibitory effects of alcohol on glucose transport across the blood-brain barrier leads to neurodegeneration: preventive role of acetyl-L: -carnitine. Psychopharmacology (Berl) 214:707-718.

Abdul Muneer PM, Alikunju S, Szlachetka AM, Haorah J (2011b) Methamphetamine inhibits the glucose uptake by human neurons and astrocytes: stabilization by acetyl-L-carnitine. PLoS One 6:e19258.

Abi-Dargham A, Kegeles LS, Zea-Ponce Y, Mawlawi O, Martinez D, Mitropoulou V, O'Flynn K, Koenigsberg HW, Van Heertum R, Cooper T, Laruelle M, Siever LJ (2004) Striatal amphetamine-induced dopamine release in patients with schizotypal personality disorder studied with single photon emission computed tomography and [123I]iodobenzamide. Biol Psychiatry 55:1001-1006.

Adachi YU, Watanabe K, Satoh T, Vizi ES (2001) Halothane potentiates the effect of methamphetamine and nomifensine on extracellular dopamine levels in rat striatum: a microdialysis study. British journal of anaesthesia 86:837-845.

Adams JD, Jr., Chang ML, Klaidman L (2001) Parkinson's disease--redox mechanisms. Curr Med Chem 8:809-814.

Adriani W, Rea M, Baviera M, Invernizzi W, Carli M, Ghirardi O, Caprioli A, Laviola G (2004) Acetyl-L-carnitine reduces impulsive behaviour in adolescent rats. Psychopharmacology (Berl) 176:296-304.

Ahmadian S, Barar J, Saei AA, Fakhree MA, Omidi Y (2009) Cellular toxicity of nanogenomedicine in MCF-7 cell line: MTT assay. Journal of visualized experiments : JoVE.

Ahmed HH (2012) Modulatory effects of vitamin E, acetyl-L-carnitine and alpha-lipoic acid on new potential biomarkers for Alzheimer's disease in rat model. Exp Toxicol Pathol 64:549-556.

Akiyama K, Kanzaki A, Tsuchida K, Ujike H (1994) Methamphetamine-induced behavioral sensitization and its implications for relapse of schizophrenia. Schizophrenia research 12:251-257.

Ali SF, Newport GD, Holson RR, Slikker W, Jr., Bowyer JF (1994) Low environmental temperatures or pharmacologic agents that produce hypothermia decrease methamphetamine neurotoxicity in mice. Brain Res 658:33-38.

Almeida A, Almeida J, Bolanos JP, Moncada S (2001) Different responses of astrocytes and neurons to nitric oxide: the role of glycolytically generated ATP in astrocyte protection. Proc Natl Acad Sci U S A 98:15294-15299.

Aloj L, Caraco C, Jagoda E, Eckelman WC, Neumann RD (1999) Glut-1 and hexokinase expression: relationship with 2-fluoro-2-deoxy-D-glucose uptake in A431 and T47D cells in culture. Cancer Res 59:4709-4714.

Alves E, Binienda Z, Carvalho F, Alves CJ, Fernandes E, de Lourdes Bastos M, Tavares MA, Summavielle T (2009) Acetyl-L-carnitine provides effective in vivo neuroprotection over 3,4-methylenedioximethamphetamine-induced mitochondrial neurotoxicity in the adolescent rat brain. Neuroscience 158:514-523.

Angelini C, Vergani L, Martinuzzi A (1992) Clinical and biochemical aspects of carnitine deficiency and insufficiency: transport defects and inborn errors of beta-oxidation. Crit Rev Clin Lab Sci 29:217-242.

Angelucci L, Ramacci MT, Taglialatela G, Hulsebosch C, Morgan B, Werrbach-Perez K, Perez-Polo R (1988) Nerve growth factor binding in aged rat central nervous system: effect of acetyl-L-carnitine. J Neurosci Res 20:491-496.

Arduini A, Denisova N, Virmani A, Avrova N, Federici G, Arrigoni-Martelli E (1994) Evidence for the involvement of carnitine-dependent long-chain acyltransferases in neuronal triglyceride and phospholipid fatty acid turnover. J Neurochem 62:1530-1538.

Arduini A, Rossi M, Mancinelli G, Belfiglio M, Scurti R, Radatti G, Shohet SB (1990) Effect of L-carnitine and acetyl-L-carnitine on the human erythrocyte membrane stability and deformability. Life Sci 47:2395-2400.

Armstrong BD, Noguchi KK (2004) The neurotoxic effects of 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine on serotonin, dopamine, and GABA-ergic terminals: an in-vitro autoradiographic study in rats. Neurotoxicology 25:905-914.

Page 124: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

124

Aureli T, Miccheli A, Di Cocco ME, Ghirardi O, Giuliani A, Ramacci MT, Conti F (1994) Effect of acetyl-L-carnitine on recovery of brain phosphorus metabolites and lactic acid level during reperfusion after cerebral ischemia in the rat--study by 13P- and 1H-NMR spectroscopy. Brain Res 643:92-99.

Aureli T, Di Cocco ME, Capuani G, Ricciolini R, Manetti C, Miccheli A, Conti F (2000) Effect of long-term feeding with acetyl-L-carnitine on the age-related changes in rat brain lipid composition: a study by 31P NMR spectroscopy. Neurochem Res 25:395-399.

Aureli T, Di Cocco ME, Puccetti C, Ricciolini R, Scalibastri M, Miccheli A, Manetti C, Conti F (1998) Acetyl-L-carnitine modulates glucose metabolism and stimulates glycogen synthesis in rat brain. Brain Res 796:75-81.

Aureli T, Puccetti C, Di Cocco ME, Arduini A, Ricciolini R, Scalibastri M, Manetti C, Conti F (1999) Entry of [(1,2-13C2)acetyl]-L-carnitine in liver tricarboxylic acid cycle and lipogenesis: a study by 13C NMR spectroscopy in conscious, freely moving rats. Eur J Biochem 263:287-293.

Bae BI, Xu H, Igarashi S, Fujimuro M, Agrawal N, Taya Y, Hayward SD, Moran TH, Montell C, Ross CA, Snyder SH, Sawa A (2005) p53 mediates cellular dysfunction and behavioral abnormalities in Huntington's disease. Neuron 47:29-41.

Barr AM, Panenka WJ, MacEwan GW, Thornton AE, Lang DJ, Honer WG, Lecomte T (2006) The need for speed: an update on methamphetamine addiction. Journal of psychiatry & neuroscience : JPN 31:301-313.

Baucum AJ, 2nd, Rau KS, Riddle EL, Hanson GR, Fleckenstein AE (2004) Methamphetamine increases dopamine transporter higher molecular weight complex formation via a dopamine- and hyperthermia-associated mechanism. J Neurosci 24:3436-3443.

Bellinghieri G, Santoro D, Calvani M, Mallamace A, Savica V (2003) Carnitine and hemodialysis. Am J Kidney Dis 41:S116-122.

Berezowski V, Miecz D, Marszalek M, Broer A, Broer S, Cecchelli R, Nalecz KA (2004) Involvement of OCTN2 and B0,+ in the transport of carnitine through an in vitro model of the blood-brain barrier. J Neurochem 91:860-872.

Bernardi P, Scorrano L, Colonna R, Petronilli V, Di Lisa F (1999) Mitochondria and cell death. Mechanistic aspects and methodological issues. Eur J Biochem 264:687-701.

Blass JP (1999) Mitochondria, neurodegenerative diseases, and selective neuronal vulnerability. Ann N Y Acad Sci 893:434-439.

Blass JP (2000) The mitochondrial spiral. An adequate cause of dementia in the Alzheimer's syndrome. Ann N Y Acad Sci 924:170-183.

Blass JP (2002) Glucose/mitochondria in neurological conditions. In: Int Rev Neurobiol (Donard SD, ed), pp 325-376: Academic Press.

Bondy CA, Cheng CM (2004) Signaling by insulin-like growth factor 1 in brain. Eur J Pharmacol 490:25-31.

Booij J, Kemp P (2008) Dopamine transporter imaging with [(123)I]FP-CIT SPECT: potential effects of drugs. Eur J Nucl Med Mol Imaging 35:424-438.

Booij J, de Bruin K, Gunning WB (2006) Repeated administration of D-amphetamine induces loss of [123I]FP-CIT binding to striatal dopamine transporters in rat brain: a validation study. Nucl Med Biol 33:409-411.

Bourque M, Liu B, Dluzen DE, Di Paolo T (2011) Sex differences in methamphetamine toxicity in mice: effect on brain dopamine signaling pathways. Psychoneuroendocrinology 36:955-969.

Bowyer JF (1995) The role of hyperthermia in amphetamine's interactions with NMDA receptors, nitric oxide, and age to produce neurotoxicity. Ann N Y Acad Sci 765:309-310.

Bowyer JF, Davies DL, Schmued L, Broening HW, Newport GD, Slikker W, Jr., Holson RR (1994) Further studies of the role of hyperthermia in methamphetamine neurotoxicity. J Pharmacol Exp Ther 268:1571-1580.

Boyt AA, Taddei TK, Hallmayer J, Helmerhorst E, Gandy SE, Craft S, Martins RN (2000) The effect of insulin and glucose on the plasma concentration of Alzheimer's amyloid precursor protein. Neuroscience 95:727-734.

Brass EP (2004) Carnitine and sports medicine: use or abuse? Ann N Y Acad Sci 1033:67-78. Bremer J (1962) Carnitine in intermediary metabolism. The metabolism of fatty acid esters of

carnitine by mitochondria. J Biol Chem 237:3628-3632. Bremer J (1983) Carnitine - metabolism and functions. Physiol Rev 63:1420-1480. Bremer J (1990) The role of carnitine in intracellular metabolism. J Clin Chem Clin Biochem

28:297-301.

Page 125: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

125

Broderick TL, Quinney HA, Lopaschuk GD (1992) Carnitine stimulation of glucose oxidation in the fatty acid perfused isolated working rat heart. J Biol Chem 267:3758-3763.

Broderick TL, Quinney HA, Lopaschuk GD (1995a) L-carnitine increases glucose metabolism and mechanical function following ischaemia in diabetic rat heart. Cardiovascular research 29:373-378.

Broderick TL, Driedzic W, Paulson DJ (2000) Propionyl-L-carnitine effects on postischemic recovery of heart function and substrate oxidation in the diabetic rat. Mol Cell Biochem 206:151-157.

Broderick TL, Panagakis G, DiDomenico D, Gamble J, Lopaschuk GD, Shug AL, Paulson DJ (1995b) L-carnitine improvement of cardiac function is associated with a stimulation in glucose but not fatty acid metabolism in carnitine-deficient hearts. Cardiovascular research 30:815-820.

Brotchie J, Fitzer-Attas C (2009) Mechanisms compensating for dopamine loss in early Parkinson disease. Neurology 72:S32-38.

Brown GC, Borutaite V (2011) There is no evidence that mitochondria are the main source of reactive oxygen species in mammalian cells. Mitochondrion.

Brown JM, Yamamoto BK (2003) Effects of amphetamines on mitochondrial function: role of free radicals and oxidative stress. Pharmacology & therapeutics 99:45-53.

Brown JM, Quinton MS, Yamamoto BK (2005) Methamphetamine-induced inhibition of mitochondrial complex II: roles of glutamate and peroxynitrite. J Neurochem 95:429-436.

Burrows KB, Gudelsky G, Yamamoto BK (2000) Rapid and transient inhibition of mitochondrial function following methamphetamine or 3,4-methylenedioxymethamphetamine administration. Eur J Pharmacol 398:11-18.

Cadet JL, Brannock C (1998) Free radicals and the pathobiology of brain dopamine systems. Neurochem Int 32:117-131.

Cadet JL, Jayanthi S, Deng X (2003) Speed kills: cellular and molecular bases of methamphetamine-induced nerve terminal degeneration and neuronal apoptosis. FASEB J 17:1775-1788.

Cadet JL, Krasnova IN, Jayanthi S, Lyles J (2007) Neurotoxicity of substituted amphetamines: molecular and cellular mechanisms. Neurotox Res 11:183-202.

Calabrese V, Giuffrida Stella AM, Calvani M, Butterfield DA (2006) Acetylcarnitine and cellular stress response: roles in nutritional redox homeostasis and regulation of longevity genes. J Nutr Biochem 17:73-88.

Cande C, Cecconi F, Dessen P, Kroemer G (2002) Apoptosis-inducing factor (AIF): key to the conserved caspase-independent pathways of cell death? J Cell Sci 115:4727-4734.

Capaldo B, Napoli R, Di Bonito P, Albano G, Sacca L (1991) Carnitine improves peripheral glucose disposal in non-insulin-dependent diabetic patients. Diabetes Res Clin Pract 14:191-195.

Carson RE, Breier A, de Bartolomeis A, Saunders RC, Su TP, Schmall B, Der MG, Pickar D, Eckelman WC (1997) Quantification of amphetamine-induced changes in [11C]raclopride binding with continuous infusion. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 17:437-447.

Carter AL, Abney TO, Lapp DF (1995) Biosynthesis and metabolism of carnitine. J Child Neurol 10 Suppl 2:S3-7.

Cass WA (1997) Decreases in evoked overflow of dopamine in rat striatum after neurotoxic doses of methamphetamine. J Pharmacol Exp Ther 280:105-113.

Castorina M, Ambrosini AM, Pacific L, Ramacci MT, Angelucci L (1994) Age-dependent loss of NMDA receptors in hippocampus, striatum, and frontal cortex of the rat: prevention by acetyl-L-carnitine. Neurochem Res 19:795-798.

Cheng CM, Kelley B, Wang J, Strauss D, Eagles DA, Bondy CA (2003) A ketogenic diet increases brain insulin-like growth factor receptor and glucose transporter gene expression. Endocrinology 144:2676-2682.

Chinta SJ, Mallajosyula JK, Rane A, Andersen JK (2010) Mitochondrial alpha-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo. Neurosci Lett 486:235-239.

Chu PW, Hadlock GC, Vieira-Brock P, Stout K, Hanson GR, Fleckenstein AE (2010) Methamphetamine alters vesicular monoamine transporter-2 function and potassium-stimulated dopamine release. J Neurochem 115:325-332.

Cifone MG, Alesse E, Di Marzio L, Ruggeri B, Zazzeroni F, Moretti S, Famularo G, Steinberg SM, Vullo E, De Simone C (1997) Effect of L-carnitine treatment in vivo on apoptosis and

Page 126: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

126

ceramide generation in peripheral blood lymphocytes from AIDS patients. Proc Assoc Am Physicians 109:146-153.

Coccurello R, Caprioli A, Ghirardi O, Virmani A (2007) Valproate and acetyl-L-carnitine prevent methamphetamine-induced behavioral sensitization in mice. Ann N Y Acad Sci 1122:260-275.

Coskun PE, Beal MF, Wallace DC (2004) Alzheimer's brains harbor somatic mtDNA control-region mutations that suppress mitochondrial transcription and replication. Proc Natl Acad Sci U S A 101:10726-10731.

Craft S, Asthana S, Schellenberg G, Baker L, Cherrier M, Boyt AA, Martins RN, Raskind M, Peskind E, Plymate S (2000) Insulin effects on glucose metabolism, memory, and plasma amyloid precursor protein in Alzheimer's disease differ according to apolipoprotein-E genotype. Ann N Y Acad Sci 903:222-228.

Cruz NF, Dienel GA (2002) High glycogen levels in brains of rats with minimal environmental stimuli: implications for metabolic contributions of working astrocytes. J Cereb Blood Flow Metab 22:1476-1489.

Cubells JF, Rayport S, Rajendran G, Sulzer D (1994) Methamphetamine neurotoxicity involves vacuolation of endocytic organelles and dopamine-dependent intracellular oxidative stress. J Neurosci 14:2260-2271.

Daumann J, Koester P, Becker B, Wagner D, Imperati D, Gouzoulis-Mayfrank E, Tittgemeyer M (2011) Medial prefrontal gray matter volume reductions in users of amphetamine-type stimulants revealed by combined tract-based spatial statistics and voxel-based morphometry. NeuroImage 54:794-801.

Davidson C, Gow AJ, Lee TH, Ellinwood EH (2001) Methamphetamine neurotoxicity: necrotic and apoptotic mechanisms and relevance to human abuse and treatment. Brain research Brain research reviews 36:1-22.

De Gaetano A, Mingrone G, Castagneto M, Calvani M (1999) Carnitine increases glucose disposal in humans. J Am Coll Nutr 18:289-295.

De Simone R, Ramacci MT, Aloe L (1991) Effect of acetyl-L-carnitine on forebrain cholinergic neurons of developing rats. Int J Dev Neurosci 9:39-46.

De Vito MJ, Wagner GC (1989) Methamphetamine-induced neuronal damage: a possible role for free radicals. Neuropharmacology 28:1145-1150.

Demarquoy J, Georges B, Rigault C, Royer MC, Clairet A, Soty M, Lekounoungou S, Le Borgne F (2004) Radioisotopic determination of L -carnitine content in foods commonly eaten in Western countries. Food Chem 86:137-142.

Dewey SL, Chaurasia CS, Chen CE, Volkow ND, Clarkson FA, Porter SP, Straughter-Moore RM, Alexoff DL, Tedeschi D, Russo NB, Fowler JS, Brodie JD (1997) GABAergic attenuation of cocaine-induced dopamine release and locomotor activity. Synapse 25:393-398.

Dhitavat S, Ortiz D, Rogers E, Rivera E, Shea TB (2005) Folate, vitamin E, and acetyl-L-carnitine provide synergistic protection against oxidative stress resulting from exposure of human neuroblastoma cells to amyloid-beta. Brain Res 1061:114-117.

Di Chiara G, Imperato A (1988) Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc Natl Acad Sci U S A 85:5274-5278.

DiMauro S, DiMauro PM (1973) Muscle carnitine palmityltransferase deficiency and myoglobinuria. Science 182:929-931.

DiNuzzo M, Maraviglia B, Giove F (2011) Why does the brain (not) have glycogen? Bioessays 33:319-326.

Dringen R, Wiesinger H, Hamprecht B (1993) Uptake of L-lactate by cultured rat brain neurons. Neurosci Lett 163:5-7.

Droge W (2002) Free radicals in the physiological control of cell function. Physiol Rev 82:47-95. Du C, Fang M, Li Y, Li L, Wang X (2000) Smac, a mitochondrial protein that promotes cytochrome

c-dependent caspase activation by eliminating IAP inhibition. Cell 102:33-42. Edmond J (1992) Energy metabolism in developing brain cells. Can J Physiol Pharmacol 70

Suppl:S118-129. Eisch AJ, Gaffney M, Weihmuller FB, O'Dell SJ, Marshall JF (1992) Striatal subregions are

differentially vulnerable to the neurotoxic effects of methamphetamine. Brain Res 598:321-326.

El-Awady el SE, Moustafa YM, Abo-Elmatty DM, Radwan A (2010) Cisplatin-induced cardiotoxicity: Mechanisms and cardioprotective strategies. Eur J Pharmacol 650:335-341.

Page 127: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

127

Engel AG, Angelini C (1973) Carnitine deficiency of human skeletal muscle with associated lipid storage myopathy: a new syndrome. Science 179:899-902.

Evans AM, Fornasini G (2003) Pharmacokinetics of L-carnitine. Clin Pharmacokinet 42:941-967. Falchetto S, Kato G, Provini L (1971) The action of carnitines on cortical neurons. Can J Physiol

Pharmacol 49:1-7. Famularo G, De Simone C, Tzantzoglou S, Trinchieri V (1994) Apoptosis, anti-apoptotic

compounds and TNF-alpha release. Immunol Today 15:495-496. Fariello RG, Ferraro TN, Golden GT, DeMattei M (1988) Systemic acetyl-L-carnitine elevates nigral

levels of glutathione and GABA. Life Sci 43:289-292. Farrell S, Vogel J, Bieber LL (1986) Entry of acetyl-L-carnitine into biosynthetic pathways. Biochim

Biophys Acta 876:175-177. Felix C, Gillis M, Driedzic WR, Paulson DJ, Broderick TL (2001) Effects of propionyl-L-carnitine on

isolated mitochondrial function in the reperfused diabetic rat heart. Diabetes Res Clin Pract 53:17-24.

Fernandez AM, Torres-Aleman I (2012) The many faces of insulin-like peptide signalling in the brain. Nature reviews Neuroscience 13:225-239.

Ferrannini E, Buzzigoli G, Bevilacqua S, Boni C, Del Chiaro D, Oleggini M, Brandi L, Maccari F (1988) Interaction of carnitine with insulin-stimulated glucose metabolism in humans. Am J Physiol 255:E946-952.

Ferrucci M, Pasquali L, Paparelli A, Ruggieri S, Fornai F (2008a) Pathways of methamphetamine toxicity. Ann N Y Acad Sci 1139:177-185.

Ferrucci M, Pasquali L, Ruggieri S, Paparelli A, Fornai F (2008b) Alpha-synuclein and autophagy as common steps in neurodegeneration. Parkinsonism & related disorders 14 Suppl 2:S180-184.

Filosto M, Scarpelli M, Cotelli MS, Vielmi V, Todeschini A, Gregorelli V, Tonin P, Tomelleri G, Padovani A (2011) The role of mitochondria in neurodegenerative diseases. Journal of neurology 258:1763-1774.

Finn PF, Dice JF (2006) Proteolytic and lipolytic responses to starvation. Nutrition 22:830-844. Flanagan JL, Simmons PA, Vehige J, Willcox MDP, Garrett Q (2010) Role of carnitine in disease.

Nutrition & Metabolism 7. Fleckenstein AE, Volz TJ, Riddle EL, Gibb JW, Hanson GR (2007) New insights into the

mechanism of action of amphetamines. Annual review of pharmacology and toxicology 47:681-698.

Fornai F, Lenzi P, Capobianco L, Iacovelli L, Scarselli P, Lazzeri G, De Blasi A (2008) Involvement of dopamine receptors and beta-arrestin in metamphetamine-induced inclusions formation in PC12 cells. J Neurochem 105:1939-1947.

Fornai F, Lenzi P, Lazzeri G, Ferrucci M, Fulceri F, Giorgi FS, Falleni A, Ruggieri S, Paparelli A (2007) Fine ultrastructure and biochemistry of PC12 cells: a comparative approach to understand neurotoxicity. Brain Res 1129:174-190.

Fornai F, Lenzi P, Gesi M, Soldani P, Ferrucci M, Lazzeri G, Capobianco L, Battaglia G, De Blasi A, Nicoletti F, Paparelli A (2004) Methamphetamine produces neuronal inclusions in the nigrostriatal system and in PC12 cells. J Neurochem 88:114-123.

Franc BL, Acton PD, Mari C, Hasegawa BH (2008) Small-animal SPECT and SPECT/CT: important tools for preclinical investigation. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 49:1651-1663.

Friedman S, Fraenkel G (1955) Reversible enzymatic acetylation of carnitine. Arch Biochem Biophys 59:491-501.

Fritz I (1955) The effect of muscle extracts on the oxidation of palmitic acid by liver slices and homogenates. Acta Physiol Scand 34:367-385.

Frohlich J, Seccombe DW, Hahn P, Dodek P, Hynie I (1978) Effect of fasting on free and esterified carnitine levels in human serum and urine: correlation with serum levels of free fatty acids and beta-hydroxybutyrate. Metabolism 27:555-561.

Frolich L, Blum-Degen D, Riederer P, Hoyer S (1999) A disturbance in the neuronal insulin receptor signal transduction in sporadic Alzheimer's disease. Ann N Y Acad Sci 893:290-293.

Fukumura M, Cappon GD, Pu C, Broening HW, Vorhees CV (1998) A single dose model of methamphetamine-induced neurotoxicity in rats: effects on neostriatal monoamines and glial fibrillary acidic protein. Brain Res 806:1-7.

Page 128: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

128

Fumagalli F, Gainetdinov RR, Valenzano KJ, Caron MG (1998) Role of dopamine transporter in methamphetamine-induced neurotoxicity: evidence from mice lacking the transporter. J Neurosci 18:4861-4869.

Gadaleta MN, Petruzzella V, Renis M, Fracasso F, Cantatore P (1990) Reduced transcription of mitochondrial DNA in the senescent rat. Tissue dependence and effect of L-carnitine. Eur J Biochem 187:501-506.

Gajkowska B, Motyl T, Olszewska-Badarczuk H, Godlewski MM (2001) Expression of BAX in cell nucleus after experimentally induced apoptosis revealed by immunogold and embedment-free electron microscopy. Cell biology international 25:725-733.

Gandhi S, Abramov AY (2012) Mechanism of oxidative stress in neurodegeneration. Oxidative medicine and cellular longevity 2012:428010.

Gehrmann J, Yao DL, Bonetti B, Bondy CA, Brenner M, Zhou J, Kreutzberg GW, Webster HD (1994) Expression of insulin-like growth factor-I and related peptides during motoneuron regeneration. Exp Neurol 128:202-210.

Genead R, Fischer H, Hussain A, Jaksch M, Andersson AB, Ljung K, Bulatovic I, Franco-Cereceda A, Elsheikh E, Corbascio M, Smith CI, Sylven C, Grinnemo KH (2012) Ischemia-reperfusion injury and pregnancy initiate time-dependent and robust signs of up-regulation of cardiac progenitor cells. PLoS One 7:e36804.

Genuth SM, Hoppel CL (1979) Plasma and urine carnitine in diabetic ketosis. Diabetes 28:1083-1087.

Gerozissis K, Rouch C, Lemierre S, Nicolaidis S, Orosco M (2001) A potential role of central insulin in learning and memory related to feeding. Cell Mol Neurobiol 21:389-401.

Gibb JW, Kogan FJ (1979) Influence of dopamine synthesis on methamphetamine-induced changes in striatal and adrenal tyrosine hydroxylase activity. Naunyn-Schmiedeberg's archives of pharmacology 310:185-187.

Gibson GE, Blass JP, Beal MF, Bunik V (2005) The alpha-ketoglutarate-dehydrogenase complex: a mediator between mitochondria and oxidative stress in neurodegeneration. Mol Neurobiol 31:43-63.

Gill MB, Bockhorst K, Narayana P, Perez-Polo JR (2008) Bax shuttling after neonatal hypoxia-ischemia: hyperoxia effects. J Neurosci Res 86:3584-3604.

Ginovart N (2005) Imaging the dopamine system with in vivo [11C]raclopride displacement studies: understanding the true mechanism. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging 7:45-52.

Gluck MR, Moy LY, Jayatilleke E, Hogan KA, Manzino L, Sonsalla PK (2001) Parallel increases in lipid and protein oxidative markers in several mouse brain regions after methamphetamine treatment. J Neurochem 79:152-160.

Gonçalves J, Martins T, Ferreira R, Milhazes N, Borges F, Ribeiro CF, Malva JO, Macedo TR, Silva AP (2008) Methamphetamine-induced early increase of IL-6 and TNF-alpha mRNA expression in the mouse brain. Ann N Y Acad Sci 1139:103-111.

Goo MJ, Choi SM, Kim SH, Ahn BO (2012) Protective effects of acetyl-L-carnitine on neurodegenarative changes in chronic cerebral ischemia models and learning-memory impairment in aged rats. Arch Pharm Res 35:145-154.

Gorini A, Ghigini B, Villa RF (1996) Acetylcholinesterase activity of synaptic plasma membranes during ageing: effect of L-acetylcarnitine. Dementia 7:147-154.

Gorini A, D'Angelo A, Villa RF (1998) Action of L-acetylcarnitine on different cerebral mitochondrial populations from cerebral cortex. Neurochem Res 23:1485-1491.

Gouzoulis-Mayfrank E, Schreckenberger M, Sabri O, Arning C, Thelen B, Spitzer M, Kovar KA, Hermle L, Bull U, Sass H (1999) Neurometabolic effects of psilocybin, 3,4-methylenedioxyethylamphetamine (MDE) and d-methamphetamine in healthy volunteers. A double-blind, placebo-controlled PET study with [18F]FDG. Neuropsychopharmacology 20:565-581.

Greene LA, Tischler AS (1976) Establishment of a neuroadrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci U S A 73:2424-2428.

Griffith OW, Mulcahy RT (1999) The enzymes of glutathione synthesis: gamma-glutamylcysteine synthetase. Advances in enzymology and related areas of molecular biology 73:209-267, xii.

Gross CJ, Henderson LM, Savaiano DA (1986) Uptake of L-carnitine, D-carnitine and acetyl-L-carnitine by isolated guinea-pig enterocytes. Biochim Biophys Acta 886:425-433.

Page 129: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

129

Gross NB, Duncker PC, Marshall JF (2011) Striatal dopamine D1 and D2 receptors: widespread influences on methamphetamine-induced dopamine and serotonin neurotoxicity. Synapse 65:1144-1155.

Gulewitsch VS, Krimberg R (1905) Zur Kenntnis der Extraktionsstoffe der Muskeln. Z Physiol Chem 45:326-330.

Han M, Im DS (2008) Effects of mitochondrial inhibitors on cell viability in U937 monocytes under glucose deprivation. Arch Pharm Res 31:749-757.

Hao J, Shen W, Sun L, Long J, Sharman E, Shi X, Liu J (2011) Mitochondrial dysfunction in the liver of type 2 diabetic Goto-Kakizaki rats: improvement by a combination of nutrients. Br J Nutr 106:648-655.

Haorah J, Floreani NA, Knipe B, Persidsky Y (2011) Stabilization of superoxide dismutase by acetyl-l-carnitine in human brain endothelium during alcohol exposure: novel protective approach. Free Radic Biol Med 51:1601-1609.

Harold C, Wallace T, Friedman R, Gudelsky G, Yamamoto B (2000) Methamphetamine selectively alters brain glutathione. Eur J Pharmacol 400:99-102.

Harper P, Wadstrom C, Cederblad G (1993) Carnitine measurements in liver, muscle tissue, and blood in normal subjects. Clin Chem 39:592-599.

Harris MH, Thompson CB (2000) The role of the Bcl-2 family in the regulation of outer mitochondrial membrane permeability. Cell death and differentiation 7:1182-1191.

Heininger K (1999) A unifying hypothesis of Alzheimer's disease. I. Ageing sets the stage. Human Psychopharmacology: Clinical and Experimental 14:363-414.

Heininger K (2002a) The cerebral glucose-fatty acid cycle: evolutionary roots, regulation, and (patho)physiological importance. International review of neurobiology 51:103-158.

Heininger K (2002b) Aging is a deprivation syndrome driven by a germ-soma conflict. Ageing research reviews 1:481-536.

Henriksen G, Drzezga A (2011) Imaging in neurology research II: PET imaging in CNS disorders. In: Small Animal Imaging (Kiessling F, Pichler BJ, eds), pp 499-513. Heidelberg: Springer-Verlag.

Heo YR, Kang CW, Cha YS (2001) L-Carnitine changes the levels of insulin-like growth factors (IGFs) and IGF binding proteins in streptozotocin-induced diabetic rat. J Nutr Sci Vitaminol 47:329-334.

Hernandez L, Lee F, Hoebel BG (1987) Simultaneous microdialysis and amphetamine infusion in the nucleus accumbens and striatum of freely moving rats: increase in extracellular dopamine and serotonin. Brain research bulletin 19:623-628.

Hertz L, Dienel GA (2002) Energy metabolism in the brain. International review of neurobiology 51:1-102.

Hiatt WR, Regensteiner JG, Wolfel EE, Ruff L, Brass EP (1989) Carnitine and acylcarnitine metabolism during exercise in humans. Dependence on skeletal muscle metabolic state. J Clin Invest 84:1167-1173.

Honer M, Bruhlmeier M, Missimer J, Schubiger AP, Ametamey SM (2004) Dynamic imaging of striatal D2 receptors in mice using quad-HIDAC PET. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 45:464-470.

Hotta K, Kuwajima M, Ono A, Uenaka R, Nakajima H, Miyagawa J, Namba M, Hanafusa T, Horiuchi M, Nikaido H, Hayakawa J, Kono N, Saheki T, Matsuzawa Y (1996) Altered expression of carnitine palmitoyltransferase II in liver, muscle, and heart of mouse strain with juvenile visceral steatosis. Biochim Biophys Acta 1289:131-135.

Hsu YT, Youle RJ (1997) Nonionic detergents induce dimerization among members of the Bcl-2 family. J Biol Chem 272:13829-13834.

Hu MT, Taylor-Robinson SD, Chaudhuri KR, Bell JD, Labbe C, Cunningham VJ, Koepp MJ, Hammers A, Morris RG, Turjanski N, Brooks DJ (2000) Cortical dysfunction in non-demented Parkinson's disease patients: a combined (31)P-MRS and (18)FDG-PET study. Brain : a journal of neurology 123 ( Pt 2):340-352.

Huang YH, Tsai SJ, Su TW, Sim CB (1999) Effects of repeated high-dose methamphetamine on local cerebral glucose utilization in rats. Neuropsychopharmacology 21:427-434.

Hülsmann WC, Dubelaar ML, Lamers JM, Maccari F (1985) Protection by acyl-carnitines and phenylmethylsulfonyl fluoride of rat heart subjected to ischemia and reperfusion. Biochim Biophys Acta 847:62-66.

Hume SP, Gunn RN, Jones T (1998) Pharmacological constraints associated with positron emission tomographic scanning of small laboratory animals. European journal of nuclear medicine 25:173-176.

Page 130: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

130

Imam SZ, Oetinger M, Skinner J, Slikker W, Jr., Ali SF (2003) The role of caspase III inhibition in methamphetamine-induced alterations in p53 and bcl-2 expression: correlation with dopaminergic neurotoxicity. Ann N Y Acad Sci 993:350; discussion 387-393.

Imam SZ, Itzhak Y, Cadet JL, Islam F, Slikker W, Jr., Ali SF (2001a) Methamphetamine-induced alteration in striatal p53 and bcl-2 expressions in mice. Brain Res Mol Brain Res 91:174-178.

Imam SZ, el-Yazal J, Newport GD, Itzhak Y, Cadet JL, Slikker W, Jr., Ali SF (2001b) Methamphetamine-induced dopaminergic neurotoxicity: role of peroxynitrite and neuroprotective role of antioxidants and peroxynitrite decomposition catalysts. Ann N Y Acad Sci 939:366-380.

Imperato A, Ramacci MT, Angelucci L (1989) Acetyl-L-carnitine enhances acetylcholine release in the striatum and hippocampus of awake freely moving rats. Neurosci Lett 107:251-255.

Indiveri C, Pochini L, Oppedisano F, Tonazzi A (2010) The Carnitine Transporter Network: Interactions with Drugs. Current chemical biology 4:108-123.

Itzhak Y, Achat-Mendes C (2004) Methamphetamine and MDMA (ecstasy) neurotoxicity: 'of mice and men'. IUBMB Life 56:249-255.

Jayanthi S, Deng X, Noailles PA, Ladenheim B, Cadet JL (2004) Methamphetamine induces neuronal apoptosis via cross-talks between endoplasmic reticulum and mitochondria-dependent death cascades. FASEB J 18:238-251.

Jin K, Mao XO, Eshoo MW, Nagayama T, Minami M, Simon RP, Greenberg DA (2001) Microarray analysis of hippocampal gene expression in global cerebral ischemia. Ann Neurol 50:93-103.

Jones LL, McDonald DA, Borum PR (2010) Acylcarnitines: role in brain. Prog Lipid Res 49:61-75. Jongen C, de Bruin K, Beekman F, Booij J (2008) SPECT imaging of D2 dopamine receptors and

endogenous dopamine release in mice. Eur J Nucl Med Mol Imaging 35:1692-1698. Kang N, Ma JH, Zhou X, Fan XB, Shang XJ, Huang YF (2011) [Effects of L-carnitine on the

apoptosis of spermatogenic cells and epididymal sperm count and motility in rats with diabetes mellitus]. Zhonghua Nan Ke Xue 17:422-426.

Keller J, Ringseis R, Priebe S, Guthke R, Kluge H, Eder K (2011) Effect of L-carnitine on the hepatic transcript profile in piglets as animal model. Nutr Metab (Lond) 8:76.

Kerdsan W, Thanoi S, Nudmamud-Thanoi S (2012) Changes in the neuronal glutamate transporter EAAT3 in rat brain after exposure to methamphetamine. Basic Clin Pharmacol Toxicol 111:275-278.

Kerner J, Hoppel C (2000) Fatty acid import into mitochondria. Biochim Biophys Acta 1486:1-17. Kido Y, Tamai I, Ohnari A, Sai Y, Kagami T, Nezu J, Nikaido H, Hashimoto N, Asano M, Tsuji A

(2001) Functional relevance of carnitine transporter OCTN2 to brain distribution of L-carnitine and acetyl-L-carnitine across the blood-brain barrier. J Neurochem 79:959-969.

Kim WH, Park WB, Gao B, Jung MH (2004) Critical role of reactive oxygen species and mitochondrial membrane potential in Korean mistletoe lectin-induced apoptosis in human hepatocarcinoma cells. Mol Pharmacol 66:1383-1396.

Kish SJ, Fitzmaurice PS, Boileau I, Schmunk GA, Ang LC, Furukawa Y, Chang LJ, Wickham DJ, Sherwin A, Tong J (2009) Brain serotonin transporter in human methamphetamine users. Psychopharmacology (Berl) 202:649-661.

Kiyatkin EA, Sharma HS (2009) Acute methamphetamine intoxication: brain hyperthermia, blood-brain barrier, brain edema, and morphological cell abnormalities. International review of neurobiology 88:65-100.

Kobayashi K, Inoue O, Watanabe Y, Onoe H, Langstrom B (1995) Difference in response of D2 receptor binding between 11C-N-methylspiperone and 11C-raclopride against anesthetics in rhesus monkey brain. Journal of neural transmission General section 100:147-151.

Kobayashi S, Iwamoto M, Kon K, Waki H, Ando S, Tanaka Y (2010) Acetyl-L-carnitine improves aged brain function. Geriatr Gerontol Int 10 Suppl 1:S99-106.

Kohler C, Fuxe K, Ross SB (1981) Regional in vivo binding of [3H]N-propylnorapomorphine in the mouse brain. Evidence for labelling of central dopamine receptors. Eur J Pharmacol 72:397-402.

Koo U, Nam KW, Ham A, Lyu D, Kim B, Lee SJ, Kim KH, Oh KB, Mar W, Shin J (2011) Neuroprotective effects of 3alpha-acetoxyeudesma-1,4(15),11(13)-trien-12,6alpha-olide against dopamine-induced apoptosis in the human neuroblastoma SH-SY5Y cell line. Neurochem Res 36:1991-2001.

Kousik SM, Napier TC, Carvey PM (2012) The effects of psychostimulant drugs on blood brain barrier function and neuroinflammation. Frontiers in pharmacology 3:121.

Page 131: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

131

Krasnova IN, Justinova Z, Ladenheim B, Jayanthi S, McCoy MT, Barnes C, Warner JE, Goldberg SR, Cadet JL (2010) Methamphetamine self-administration is associated with persistent biochemical alterations in striatal and cortical dopaminergic terminals in the rat. PLoS One 5:e8790.

Kuczenski R, Segal DS, Cho AK, Melega W (1995) Hippocampus norepinephrine, caudate dopamine and serotonin, and behavioral responses to the stereoisomers of amphetamine and methamphetamine. J Neurosci 15:1308-1317.

Kung HF, Guo YZ, Billings J, Xu X, Mach RH, Blau M, Ackerhalt RE (1988) Preparation and biodistribution of [125I]IBZM: a potential CNS D-2 dopamine receptor imaging agent. International journal of radiation applications and instrumentation Part B, Nuclear medicine and biology 15:195-201.

Kung HF, Pan S, Kung MP, Billings J, Kasliwal R, Reilley J, Alavi A (1989) In vitro and in vivo evaluation of [123I]IBZM: a potential CNS D-2 dopamine receptor imaging agent. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 30:88-92.

Kunikowska G, Jenner P (2002) The distribution of copper, zinc- and manganese-superoxide dismutase, and glutathione peroxidase messenger ribonucleic acid in rat basal ganglia. Biochem Pharmacol 63:1159-1164.

Kurachi M (2003) Pathogenesis of schizophrenia: Part II. Temporo-frontal two-step hypothesis. Psychiatry and clinical neurosciences 57:9-15.

Lango R, Smolenski RT, Rogowski J, Siebert J, Wujtewicz M, Slominska EM, Lysiak-Szydlowska W, Yacoub MH (2005) Propionyl-L-carnitine improves hemodynamics and metabolic markers of cardiac perfusion during coronary surgery in diabetic patients. Cardiovasc Drugs Ther 19:267-275.

Laruelle M, Abi-Dargham A, van Dyck CH, Rosenblatt W, Zea-Ponce Y, Zoghbi SS, Baldwin RM, Charney DS, Hoffer PB, Kung HF, et al. (1995) SPECT imaging of striatal dopamine release after amphetamine challenge. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 36:1182-1190.

Laschi R, Badiali de Giorgi L, Bonvicini F, Centurione L (1990) Ultrastructural aspects of aging rat hippocampus after long-term administration of acetyl-L-carnitine. Int J Clin Pharmacol Res 10:59-63.

Lazzeri G, Lenzi P, Busceti CL, Ferrucci M, Falleni A, Bruno V, Paparelli A, Fornai F (2007) Mechanisms involved in the formation of dopamine-induced intracellular bodies within striatal neurons. J Neurochem 101:1414-1427.

Lee CH, Yin PH, Lu CY, Chi CS, Wei YH (2000) Increase of mitochondria and mitochondrial DNA in response to oxidative stress in human cells. Biochem J 348:425-432.

Lee JH, Won YS, Park KH, Lee MK, Tachibana H, Yamada K, Seo KI (2012) Celastrol inhibits growth and induces apoptotic cell death in melanoma cells via the activation ROS-dependent mitochondrial pathway and the suppression of PI3K/AKT signaling. Apoptosis.

Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443:787-795.

Liu J, Atamna H, Kuratsune H, Ames BN (2002) Delaying brain mitochondrial decay and aging with mitochondrial antioxidants and metabolites. Ann N Y Acad Sci 959:133-166.

Lohninger A, Auer B, Lechner S, Salzer H (1996) Prenatal effects of betamethasone-L-carnitine combinations on fetal rat lung. J Perinat Med 24:591-599.

London ED, Berman SM, Voytek B, Simon SL, Mandelkern MA, Monterosso J, Thompson PM, Brody AL, Geaga JA, Hong MS, Hayashi KM, Rawson RA, Ling W (2005) Cerebral metabolic dysfunction and impaired vigilance in recently abstinent methamphetamine abusers. Biol Psychiatry 58:770-778.

MacKenzie-Graham A, Lee E-F, Dinov ID, Bota M, Shattuck DW, Ruffins S, Yuan H, Konstantinidis F, Pitiot A, Ding Y, Hu G, Jacobs RE, Toga AW (2004) A multimodal, multidimensional atlas of the C57BL/6J mouse brain. Journal of anatomy 204:93-102.

Magder S (2006) Reactive oxygen species: toxic molecules or spark of life? Crit Care 10:208. Magistretti PJ, Sorg O, Naichen Y, Pellerin L, de Rham S, Martin JL (1994) Regulation of astrocyte

energy metabolism by neurotransmitters. Renal physiology and biochemistry 17:168-171. Maher F, Vannucci SJ, Simpson IA (1994) Glucose transporter proteins in brain. FASEB J 8:1003-

1011. Mandell AJ, Morgan M (1970) Amphetamine induced increase in tyrosine hydroxylase activity.

Nature 227:75-76. Mantle TJ, Tipton KF, Garrett NJ (1976) Inhibition of monoamine oxidase by amphetamine and

related compounds. Biochem Pharmacol 25:2073-2077.

Page 132: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

132

Marek GJ, Vosmer G, Seiden LS (1990a) The effects of monoamine uptake inhibitors and methamphetamine on neostriatal 6-hydroxydopamine (6-OHDA) formation, short-term monoamine depletions and locomotor activity in the rat. Brain Res 516:1-7.

Marek GJ, Vosmer G, Seiden LS (1990b) Dopamine uptake inhibitors block long-term neurotoxic effects of methamphetamine upon dopaminergic neurons. Brain Res 513:274-279.

Mark KA, Quinton MS, Russek SJ, Yamamoto BK (2007) Dynamic changes in vesicular glutamate transporter 1 function and expression related to methamphetamine-induced glutamate release. J Neurosci 27:6823-6831.

Martins T, Baptista S, Goncalves J, Leal E, Milhazes N, Borges F, Ribeiro CF, Quintela O, Lendoiro E, Lopez-Rivadulla M, Ambrosio AF, Silva AP (2011) Methamphetamine transiently increases the blood-brain barrier permeability in the hippocampus: role of tight junction proteins and matrix metalloproteinase-9. Brain Res 1411:28-40.

Mazzio E, Yoon KJ, Soliman KFA (2003a) Acetyl-L-carnitne cytoprotection against 1-methyl-4-phenylpyridinium toxicity in neuroblastoma cells. Biochemical Pharmacology 66:297-306.

Mazzio E, Yoon KJ, Soliman KF (2003b) Acetyl-L-carnitine cytoprotection against 1-methyl-4-phenylpyridinium toxicity in neuroblastoma cells. Biochem Pharmacol 66:297-306.

McKetin R, Hickey K, Devlin K, Lawrence K (2010) The risk of psychotic symptoms associated with recreational methamphetamine use. Drug and alcohol review 29:358-363.

Mingorance C, Rodriguez-Rodriguez R, Justo ML, Herrera MD, de Sotomayor MA (2011) Pharmacological effects and clinical applications of propionyl-L-carnitine. Nutr Rev 69:279-290.

Mingrone G, Greco AV, Capristo E, Benedetti G, Giancaterini A, De Gaetano A, Gasbarrini G (1999) L-carnitine improves glucose disposal in type 2 diabetic patients. J Am Coll Nutr 18:77-82.

Mirecki A, Fitzmaurice P, Ang L, Kalasinsky KS, Peretti FJ, Aiken SS, Wickham DJ, Sherwin A, Nobrega JN, Forman HJ, Kish SJ (2004) Brain antioxidant systems in human methamphetamine users. J Neurochem 89:1396-1408.

Molfino A, Cascino A, Conte C, Ramaccini C, Rossi Fanelli F, Laviano A (2010) Caloric restriction and L-carnitine administration improves insulin sensitivity in patients with impaired glucose metabolism. JPEN J Parenter Enteral Nutr 34:295-299.

Moretti JL, Hauet N, Caglar M, Rebillard O, Burak Z (2005) To use MIBI or not to use MIBI? That is the question when assessing tumour cells. Eur J Nucl Med Mol Imaging 32:836-842.

Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65:55-63.

Motyl T, Gajkowska B, Ploszaj T, Wareski P, Skierski J, Zimowska W (2000) Expression and subcellular redistribution of Bax during TGF-beta1-induced programmed cell death of HC11 mouse mammary epithelial cells. Cell Mol Biol (Noisy-le-grand) 46:175-185.

Murakami R, Tanaka A, Nakamura H (1997) The effect of starvation on brain carnitine concentration in neonatal rats. J Pediatr Gastroenterol Nutr 25:385-387.

Mutomba MC, Yuan H, Konyavko M, Adachi S, Yokoyama CB, Esser V, McGarry JD, Babior BM, Gottlieb RA (2000) Regulation of the activity of caspases by L-carnitine and palmitoylcarnitine. FEBS Lett 478:19-25.

Nagai T, Takuma K, Dohniwa M, Ibi D, Mizoguchi H, Kamei H, Nabeshima T, Yamada K (2007) Repeated methamphetamine treatment impairs spatial working memory in rats: reversal by clozapine but not haloperidol. Psychopharmacology (Berl) 194:21-32.

Nakama H, Chang L, Fein G, Shimotsu R, Jiang CS, Ernst T (2011) Methamphetamine users show greater than normal age-related cortical gray matter loss. Addiction 106:1474-1483.

Nakanishi T, Hatanaka T, Huang W, Prasad PD, Leibach FH, Ganapathy ME, Ganapathy V (2001) Na+- and Cl--coupled active transport of carnitine by the amino acid transporter ATB(0,+) from mouse colon expressed in HRPE cells and Xenopus oocytes. J Physiol 532:297-304.

Nalecz KA, Miecz D, Berezowski V, Cecchelli R (2004) Carnitine: transport and physiological functions in the brain. Mol Aspects Med 25:551-567.

Nezu J, Tamai I, Oku A, Ohashi R, Yabuuchi H, Hashimoto N, Nikaido H, Sai Y, Koizumi A, Shoji Y, Takada G, Matsuishi T, Yoshino M, Kato H, Ohura T, Tsujimoto G, Hayakawa J, Shimane M, Tsuji A (1999) Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter. Nature genetics 21:91-94.

Nikolaus S, Beu M, Wirrwar A, Antke C, Muller HW (2005) In vivo snapshots of the dopaminergic synapse in small animals. Mol Psychiatry 10:516-518.

Page 133: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

133

Nishijima T, Piriz J, Duflot S, Fernandez AM, Gaitan G, Gomez-Pinedo U, Verdugo JM, Leroy F, Soya H, Nunez A, Torres-Aleman I (2010) Neuronal activity drives localized blood-brain-barrier transport of serum insulin-like growth factor-I into the CNS. Neuron 67:834-846.

Nunomura A, Honda K, Takeda A, Hirai K, Zhu X, Smith MA, Perry G (2006) Oxidative damage to RNA in neurodegenerative diseases. Journal of biomedicine & biotechnology 2006:82323.

O'Callaghan JP, Miller DB (1994) Neurotoxicity profiles of substituted amphetamines in the C57BL/6J mouse. Journal of Pharmacology and Experimental Therapeutics 270:741-751.

Obici S, Feng Z, Arduini A, Conti R, Rossetti L (2003) Inhibition of hypothalamic carnitine palmitoyltransferase-1 decreases food intake and glucose production. Nat Med 9:756-761.

Ohsawa I, Nishimaki K, Yasuda C, Kamino K, Ohta S (2003) Deficiency in a mitochondrial aldehyde dehydrogenase increases vulnerability to oxidative stress in PC12 cells. J Neurochem 84:1110-1117.

Orikabe L, Yamasue H, Inoue H, Takayanagi Y, Mozue Y, Sudo Y, Ishii T, Itokawa M, Suzuki M, Kurachi M, Okazaki Y, Kasai K (2011) Reduced amygdala and hippocampal volumes in patients with methamphetamine psychosis. Schizophrenia research 132:183-189.

Oshibuchi H, Inada K, Sugawara H, Ishigooka J (2009) Aripiprazole and haloperidol suppress excessive dopamine release in the amygdala in response to conditioned fear stress, but show contrasting effects on basal dopamine release in methamphetamine-sensitized rats. Eur J Pharmacol 615:83-90.

Owen L, Sunram-Lea SI (2011) Metabolic agents that enhance ATP can improve cognitive functioning: a review of the evidence for glucose, oxygen, pyruvate, creatine, and L-carnitine. Nutrients 3:735-755.

Palmero S, Bottazzi C, Costa M, Leone M, Fugassa E (2000) Metabolic effects of L-carnitine on prepubertal rat Sertoli cells. Horm Metab Res 32:87-90.

Pande SV, Blanchaer MC (1970) Preferential loss of ATP-dependent long-chain fatty acid activating enzyme in mitochondria prepared using Nagarse. Biochim Biophys Acta 202:43-48.

Paradies G, Ruggiero FM, Gadaleta MN, Quagliariello E (1992) The effect of aging and acetyl-L-carnitine on the activity of the phosphate carrier and on the phospholipid composition in rat heart mitochondria. Biochim Biophys Acta 1103:324-326.

Parnetti L, Gaiti A, Mecocci P, Cadini D, Senin U (1992) Pharmacokinetics of IV and oral acetyl-L-carnitine in a multiple dose regimen in patients with senile dementia of Alzheimer type. Eur J Clin Pharmacol 42:89-93.

Pastorino JG, Snyder JW, Serroni A, Hoek JB, Farber JL (1993) Cyclosporin and carnitine prevent the anoxic death of cultured hepatocytes by inhibiting the mitochondrial permeability transition. J Biol Chem 268:13791-13798.

Patel SP, Sullivan PG, Lyttle TS, Magnuson DS, Rabchevsky AG (2012) Acetyl-L-carnitine treatment following spinal cord injury improves mitochondrial function correlated with remarkable tissue sparing and functional recovery. Neuroscience 210:296-307.

Pellerin L, Magistretti PJ (1994) Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U S A 91:10625-10629.

Pendergrass W, Wolf N, Poot M (2004) Efficacy of MitoTracker Green™ and CMXrosamine to measure changes in mitochondrial membrane potentials in living cells and tissues. Cytometry Part A 61A:162-169.

Pendyala G, Buescher JL, Fox HS (2012) Methamphetamine and inflammatory cytokines increase neuronal Na+/K+-ATPase isoform 3: relevance for HIV associated neurocognitive disorders. PLoS One 7:e37604.

Pereira C, Santos MS, Oliveira C (1998) Mitochondrial function impairment induced by amyloid beta-peptide on PC12 cells. Neuroreport 9:1749-1755.

Pereira FC, Cunha-Oliveira T, Viana SD, Travassos AS, Nunes S, Silva C, Prediger RD, Rego AC, Ali SF, Ribeiro CA (2012) Disruption of striatal glutamatergic/GABAergic homeostasis following acute methamphetamine in mice. Neurotoxicology and teratology 34:522-529.

Perumal AS, Gopal VB, Tordzro WK, Cooper TB, Cadet JL (1992) Vitamin E attenuates the toxic effects of 6-hydroxydopamine on free radical scavenging systems in rat brain. Brain research bulletin 29:699-701.

Petrozzi L, Ricci G, Giglioli NJ, Siciliano G, Mancuso M (2007) Mitochondria and neurodegeneration. Bioscience reports 27:87-104.

Page 134: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

134

Pettegrew JW, Levine J, McClure RJ (2000) Acetyl-L-carnitine physical-chemical, metabolic, and therapeutic properties: relevance for its mode of action in Alzheimer's disease and geriatric depression. Mol Psychiatry 5:616-632.

Piga R, Saito Y, Chen Z, Yoshida Y, Niki E (2005) Characterization of monochloramine toxicity on PC12 cells and protective effect of tocopherol via antioxidative function. Arch Biochem Biophys 436:101-109.

Piwnica-Worms D, Kronauge JF, Chiu ML (1990) Uptake and retention of hexakis (2-methoxyisobutyl isonitrile) technetium(I) in cultured chick myocardial cells. Mitochondrial and plasma membrane potential dependence. Circulation 82:1826-1838.

Pons R, De Vivo DC (1995) Primary and secondary carnitine deficiency syndromes. J Child Neurol 10 Suppl 2:S8-24.

Povlock SL, Schenk JO (1997) A multisubstrate kinetic mechanism of dopamine transport in the nucleus accumbens and its inhibition by cocaine. J Neurochem 69:1093-1105.

Pu C, Fisher JE, Cappon GD, Vorhees CV (1994) The effects of amfonelic acid, a dopamine uptake inhibitor, on methamphetamine-induced dopaminergic terminal degeneration and astrocytic response in rat striatum. Brain Res 649:217-224.

Ramirez SH, Potula R, Fan S, Eidem T, Papugani A, Reichenbach N, Dykstra H, Weksler BB, Romero IA, Couraud PO, Persidsky Y (2009) Methamphetamine disrupts blood-brain barrier function by induction of oxidative stress in brain endothelial cells. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 29:1933-1945.

Ramsay RR, Arduini A (1993) The carnitine acyltransferases and their role in modulating acyl-CoA pools. Arch Biochem Biophys 302:307-314.

Ramsay RR, Gandour RD, van der Leij FR (2001) Molecular enzymology of carnitine transfer and transport. Biochim Biophys Acta 1546:21-43.

Randle PJ (1998) Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev 14:263-283.

Randle PJ, Newsholme EA, Garland PB (1964) Regulation of glucose uptake by muscle. 8. Effects of fatty acids, ketone bodies and pyruvate, and of alloxan-diabetes and starvation, on the uptake and metabolic fate of glucose in rat heart and diaphragm muscles. Biochem J 93:652-665.

Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785-789.

Randle PJ, Sugden PH, Kerbey AL, Radcliffe PM, Hutson NJ (1978) Regulation of pyruvate oxidation and the conservation of glucose. Biochemical Society symposium:47-67.

Rebouche CJ (1992) Carnitine function and requirements during the life cycle. FASEB J 6:3379-3386.

Rebouche CJ (2004) Kinetics, pharmacokinetics, and regulation of L-carnitine and acetyl-L-carnitine metabolism. Ann N Y Acad Sci 1033:30-41.

Rebouche CJ (2005) Carnitine. In: Modern nutrition in health and disease, 10 Edition (Shils ME, Shike M, Ross AC, Caballero B, Cousins RJ, eds), pp 537-544. USA: Lippincott Williams & Wilkins.

Rebouche CJ, Engel AG (1984) Kinetic compartmental analysis of carnitine metabolism in the human carnitine deficiency syndromes. Evidence for alterations in tissue carnitine transport. J Clin Invest 73:857-867.

Rebouche CJ, Chenard CA (1991) Metabolic fate of dietary carnitine in human adults: identification and quantification of urinary and fecal metabolites. J Nutr 121:539-546.

Rebouche CJ, Seim H (1998) Carnitine metabolism and its regulation in microorganisms and mammals. Annu Rev Nutr 18:39-61.

Reznick AZ, Kagan VE, Ramsey R, Tsuchiya M, Khwaja S, Serbinova EA, Packer L (1992) Antiradical effects in L-propionyl carnitine protection of the heart against ischemia-reperfusion injury: the possible role of iron chelation. Arch Biochem Biophys 296:394-401.

Riddle EL, Fleckenstein AE, Hanson GR (2006) Mechanisms of methamphetamine-induced dopaminergic neurotoxicity. AAPS J 8:E413-418.

Ringseis R, Keller J, Eder K (2012) Role of carnitine in the regulation of glucose homeostasis and insulin sensitivity: evidence from in vivo and in vitro studies with carnitine supplementation and carnitine deficiency. Eur J Nutr 51:1-18.

Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ (1987) Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 237:1219-1223.

Page 135: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

135

Rosca MG, Lemieux H, Hoppel CL (2009) Mitochondria in the elderly: Is acetylcarnitine a rejuvenator? Adv Drug Deliv Rev 61:1332-1342.

Ruggenenti P, Cattaneo D, Loriga G, Ledda F, Motterlini N, Gherardi G, Orisio S, Remuzzi G (2009) Ameliorating hypertension and insulin resistance in subjects at increased cardiovascular risk: effects of acetyl-L-carnitine therapy. Hypertension 54:567-574.

Rump TJ, Abdul Muneer PM, Szlachetka AM, Lamb A, Haorei C, Alikunju S, Xiong H, Keblesh J, Liu J, Zimmerman MC, Jones J, Donohue TM, Jr., Persidsky Y, Haorah J (2010) Acetyl-L-carnitine protects neuronal function from alcohol-induced oxidative damage in the brain. Free Radic Biol Med 49:1494-1504.

Rytting E, Audus KL (2005) Novel organic cation transporter 2-mediated carnitine uptake in placental choriocarcinoma (BeWo) cells. J Pharmacol Exp Ther 312:192-198.

Sampath D, Jackson GR, Werrbach-Perez K, Perez-Polo JR (1994) Effects of nerve growth factor on glutathione peroxidase and catalase in PC12 cells. J Neurochem 62:2476-2479.

Sano H, Totsuka Y, Furuuchi T, Sakai K, Koshikawa N, Shigehara R, Kobayashi M (1982) ["Methamphetamine-stereotypy and hypermotility" in rats chronically treated with reserpine--the effect of intracerebral injection of chlorpromazine]. Nihon yakurigaku zasshi Folia pharmacologica Japonica 80:113-124.

Saunders C, Ferrer JV, Shi L, Chen J, Merrill G, Lamb ME, Leeb-Lundberg LM, Carvelli L, Javitch JA, Galli A (2000) Amphetamine-induced loss of human dopamine transporter activity: an internalization-dependent and cocaine-sensitive mechanism. Proc Natl Acad Sci U S A 97:6850-6855.

Scafidi S, Racz J, Hazelton J, McKenna MC, Fiskum G (2010a) Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain. Dev Neurosci 32:480-487.

Scafidi S, Fiskum G, Lindauer SL, Bamford P, Shi D, Hopkins I, McKenna MC (2010b) Metabolism of acetyl-L-carnitine for energy and neurotransmitter synthesis in the immature rat brain. J Neurochem 114:820-831.

Scaglia F, Wang Y, Longo N (1999) Functional characterization of the carnitine transporter defective in primary carnitine deficiency. Arch Biochem Biophys 364:99-106.

Scherfler C, Scholz SW, Donnemiller E, Decristoforo C, Oberladstatter M, Stefanova N, Diederen E, Virgolini I, Poewe W, Wenning GK (2005) Evaluation of [123I]IBZM pinhole SPECT for the detection of striatal dopamine D2 receptor availability in rats. NeuroImage 24:822-831.

Schiffer WK, Volkow ND, Fowler JS, Alexoff DL, Logan J, Dewey SL (2006) Therapeutic doses of amphetamine or methylphenidate differentially increase synaptic and extracellular dopamine. Synapse 59:243-251.

Schinder AF, Olson EC, Spitzer NC, Montal M (1996) Mitochondrial dysfunction is a primary event in glutamate neurotoxicity. J Neurosci 16:6125-6133.

Schinetti ML, Rossini D, Greco R, Bertelli A (1987) Protective action of acetylcarnitine on NADPH-induced lipid peroxidation of cardiac microsomes. Drugs Exp Clin Res 13:509-515.

Schlegel J, Neff F, Piontek G (2004) Serial induction of mutations by ethylnitrosourea in PC12 cells: a new model for a phenotypical characterization of the neurotoxic response to 6-hydroxydopamine. Journal of neuroscience methods 137:215-220.

Schober ME, Ke X, Xing B, Block BP, Requena DF, McKnight R, Lane RH (2012) Traumatic brain injury increased IGF-1B mRNA and altered IGF-1 exon 5 and promoter region epigenetic characteristics in the rat pup hippocampus. J Neurotrauma 29:2075-2085.

Schonekess BO, Allard MF, Lopaschuk GD (1995) Propionyl L-carnitine improvement of hypertrophied rat heart function is associated with an increase in cardiac efficiency. Eur J Pharmacol 286:155-166.

Sharma HS, Ali SF (2006) Alterations in blood-brain barrier function by morphine and methamphetamine. Ann N Y Acad Sci 1074:198-224.

Sharma S, Black SM (2009) Carnitine homeostasis, mitochondrial function, and cardiovascular disease. Drug Discov Today Dis Mech 6:e31-e39.

Shen W, Liu K, Tian C, Yang L, Li X, Ren J, Packer L, Head E, Sharman E, Liu J (2008) Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J Cell Biochem 104:1232-1243.

Shiba T, Yamato M, Kudo W, Watanabe T, Utsumi H, Yamada K (2011) In vivo imaging of mitochondrial function in methamphetamine-treated rats. NeuroImage 57:866-872.

Shigenaga MK, Hagen TM, Ames BN (1994) Oxidative damage and mitochondrial decay in aging. Proc Natl Acad Sci U S A 91:10771-10778.

Shotbolt P, Tziortzi AC, Searle GE, Colasanti A, van der Aart J, Abanades S, Plisson C, Miller SR, Huiban M, Beaver JD, Gunn RN, Laruelle M, Rabiner EA (2012) Within-subject comparison

Page 136: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

136

of [(11)C]-(+)-PHNO and [(11)C]raclopride sensitivity to acute amphetamine challenge in healthy humans. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 32:127-136.

Simpson IA, Dwyer D, Malide D, Moley KH, Travis A, Vannucci SJ (2008) The facilitative glucose transporter GLUT3: 20 years of distinction. Am J Physiol Endocrinol Metab 295:E242-253.

Skinbjerg M, Liow JS, Seneca N, Hong J, Lu S, Thorsell A, Heilig M, Pike VW, Halldin C, Sibley DR, Innis RB (2010) D2 dopamine receptor internalization prolongs the decrease of radioligand binding after amphetamine: a PET study in a receptor internalization-deficient mouse model. NeuroImage 50:1402-1407.

Sloan JL, Mager S (1999) Cloning and functional expression of a human Na(+) and Cl(-)-dependent neutral and cationic amino acid transporter B(0+). J Biol Chem 274:23740-23745.

Smith TA (2001) The rate-limiting step for tumor [18F]fluoro-2-deoxy-D-glucose (FDG) incorporation. Nucl Med Biol 28:1-4.

Snider SE, Vunck SA, van den Oord EJ, Adkins DE, McClay JL, Beardsley PM (2012) The glial cell modulators, ibudilast and its amino analog, AV1013, attenuate methamphetamine locomotor activity and its sensitization in mice. Eur J Pharmacol 679:75-80.

Steiber A, Kerner J, Hoppel CL (2004) Carnitine: a nutritional, biosynthetic, and functional perspective. Mol Aspects Med 25:455-473.

Stephens FB, Constantin-Teodosiu D, Greenhaff PL (2007) New insights concerning the role of carnitine in the regulation of fuel metabolism in skeletal muscle. J Physiol 581:431-444.

Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL (2006a) Insulin stimulates L-carnitine accumulation in human skeletal muscle. FASEB J 20:377-379.

Stephens FB, Constantin-Teodosiu D, Laithwaite D, Simpson EJ, Greenhaff PL (2006b) An acute increase in skeletal muscle carnitine content alters fuel metabolism in resting human skeletal muscle. J Clin Endocrinol Metab 91:5013-5018.

Sulzer D (2011) How addictive drugs disrupt presynaptic dopamine neurotransmission. Neuron 69:628-649.

Sulzer D, Zecca L (2000) Intraneuronal dopamine-quinone synthesis: a review. Neurotox Res 1:181-195.

Sulzer D, Sonders MS, Poulsen NW, Galli A (2005) Mechanisms of neurotransmitter release by amphetamines: a review. Progress in neurobiology 75:406-433.

Sun W, Ginovart N, Ko F, Seeman P, Kapur S (2003) In vivo evidence for dopamine-mediated internalization of D2-receptors after amphetamine: differential findings with [3H]raclopride versus [3H]spiperone. Molecular pharmacology 63:456-462.

Suwanjang W, Phansuwan-Pujito P, Govitrapong P, Chetsawang B (2010) The protective effect of melatonin on methamphetamine-induced calpain-dependent death pathway in human neuroblastoma SH-SY5Y cultured cells. J Pineal Res 48:94-101.

Suwanjang W, Phansuwan-Pujito P, Govitrapong P, Chetsawang B (2012) Calpastatin reduces calpain and caspase activation in methamphetamine-induced toxicity in human neuroblastoma SH-SY5Y cultured cells. Neurosci Lett 526:49-53.

Tafreshi AP, jalal R, Darvishalipoor S, Sepehri H, Adeli K (2010) Investigation on the expression of IGF-I protein in insulin-resistant rat brain. Int J Endocrinol Metab 8:138-142.

Taglialatela G, Caprioli A, Giuliani A, Ghirardi O (1996) Spatial memory and NGF levels in aged rats: natural variability and effects of acetyl-L-carnitine treatment. Exp Gerontol 31:577-587.

Terziyska N, Lutz T, Kozany C, Mokranjac D, Mesecke N, Neupert W, Herrmann JM, Hell K (2005) Mia40, a novel factor for protein import into the intermembrane space of mitochondria is able to bind metal ions. FEBS Lett 579:179-184.

Thomas C, Mackey MM, Diaz AA, Cox DP (2009) Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: implications for diseases associated with iron accumulation. Redox report : communications in free radical research 14:102-108.

Thomas DM, Francescutti-Verbeem DM, Kuhn DM (2008) The newly synthesized pool of dopamine determines the severity of methamphetamine-induced neurotoxicity. J Neurochem 105:605-616.

Thompson PM, Hayashi KM, Simon SL, Geaga JA, Hong MS, Sui Y, Lee JY, Toga AW, Ling W, London ED (2004) Structural abnormalities in the brains of human subjects who use methamphetamine. J Neurosci 24:6028-6036.

Page 137: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

137

Tocharus J, Khonthun C, Chongthammakun S, Govitrapong P (2010) Melatonin attenuates methamphetamine-induced overexpression of pro-inflammatory cytokines in microglial cell lines. J Pineal Res 48:347-352.

Tolu P, Masi F, Leggio B, Scheggi S, Tagliamonte A, De Montis MG, Gambarana C (2002) Effects of long-term acetyl-L-carnitine administration in rats: I. increased dopamine output in mesocorticolimbic areas and protection toward acute stress exposure. Neuropsychopharmacology 27:410-420.

Tomita M, Sendju Y (1927) Über die oxyaminoverbindungen, welche die biuretreaktion zeigen. III.Spaltung der m-amino-b-oxybuttersäure in die optisch-aktiven komponenten. Z Physiol Chem 169:263-277.

Toth E, Harsing LG, Jr., Sershen H, Ramacci MT, Lajtha A (1993) Effect of acetyl-L-carnitine on extracellular amino acid levels in vivo in rat brain regions. Neurochem Res 18:573-578.

Treem WR, Stanley CA, Finegold DN, Hale DE, Coates PM (1988) Primary carnitine deficiency due to a failure of carnitine transport in kidney, muscle, and fibroblasts. N Engl J Med 319:1331-1336.

Tsukada H, Nishiyama S, Kakiuchi T, Ohba H, Sato K, Harada N, Nakanishi S (1999) Isoflurane anesthesia enhances the inhibitory effects of cocaine and GBR12909 on dopamine transporter: PET studies in combination with microdialysis in the monkey brain. Brain Res 849:85-96.

Tucek S (1983) Acetylcoenzyme A and the synthesis of acetylcholine in neurones: review of recent progress. General physiology and biophysics 2:313-324.

Tucek S (1990) The synthesis of acetylcholine: twenty years of progress. Progress in brain research 84:467-477.

Uehara T, Sumiyoshi T, Itoh H, Kurachi M (2004) Inhibition of dopamine synthesis with alpha-methyl-p-tyrosine abolishes the enhancement of methamphetamine-induced extracellular dopamine levels in the amygdala of rats with excitotoxic lesions of the entorhinal cortex. Neurosci Lett 356:21-24.

Uziel G, Garavaglia B, Di Donato S (1988) Carnitine stimulation of pyruvate dehydrogenase complex (PDHC) in isolated human skeletal muscle mitochondria. Muscle Nerve 11:720-724.

Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J (2007) Free radicals and antioxidants in normal physiological functions and human disease. The international journal of biochemistry & cell biology 39:44-84.

Vannucci SJ, Maher F, Simpson IA (1997) Glucose transporter proteins in brain: delivery of glucose to neurons and glia. Glia 21:2-21.

Vargiu R, Littarru GP, Faa G, Mancinelli R (2008) Positive inotropic effect of coenzyme Q10, omega-3 fatty acids and propionyl-L-carnitine on papillary muscle force-frequency responses of BIO TO-2 cardiomyopathic Syrian hamsters. Biofactors 32:135-144.

Vaz FM, van Gool S, Ofman R, L IJ, Wanders RJ (1999) Carnitine biosynthesis. Purification of gamma-butyrobetaine hydroxylase from rat liver. Adv Exp Med Biol 466:117-124.

Verhoeff NP, Brucke T, Podreka I, Bobeldijk M, Angelberger P, Van Royen EA (1991a) Dynamic SPECT in two healthy volunteers to determine the optimal time for in vivo D2 dopamine receptor imaging with 123I-IBZM using the rotating gamma camera. Nuclear medicine communications 12:687-697.

Verhoeff NP, Bobeldijk M, Feenstra MG, Boer GJ, Maas MA, Erdtsieck-Ernste E, de Bruin K, van Royen EA (1991b) In vitro and in vivo D2-dopamine receptor binding with [123I]S(-) iodobenzamide ([123I]IBZM) in rat and human brain. International journal of radiation applications and instrumentation Part B, Nuclear medicine and biology 18:837-846.

Virmani A, Binienda Z (2004) Role of carnitine esters in brain neuropathology. Molecular Aspects of Medicine 25:533-549.

Virmani A, Gaetani F, Imam S, Binienda Z, Ali S (2003) Possible mechanism for the neuroprotective effects of L-carnitine on methamphetamine-evoked neurotoxicity. Ann N Y Acad Sci 993:197-207; discussion 287-198.

Virmani A, Gaetani F, Binienda Z, Xu A, Duhart H, Ali SF (2004) Role of mitochondrial dysfunction in neurotoxicity of MPP+: partial protection of PC12 cells by acetyl-L-carnitine. Ann N Y Acad Sci 1025:267-273.

Volkow ND, Wang GJ, Fowler JS, Tomasi D, Telang F (2011) Addiction: beyond dopamine reward circuitry. Proc Natl Acad Sci U S A 108:15037-15042.

Page 138: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

138

Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding YS, Wong C, Logan J (2001a) Higher cortical and lower subcortical metabolism in detoxified methamphetamine abusers. The American journal of psychiatry 158:383-389.

Volkow ND, Chang L, Wang GJ, Fowler JS, Franceschi D, Sedler M, Gatley SJ, Miller E, Hitzemann R, Ding YS, Logan J (2001b) Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence. J Neurosci 21:9414-9418.

Volkow ND, Wang GJ, Franceschi D, Fowler JS, Thanos PP, Maynard L, Gatley SJ, Wong C, Veech RL, Kunos G, Kai Li T (2006) Low doses of alcohol substantially decrease glucose metabolism in the human brain. NeuroImage 29:295-301.

Volkow ND, Kim SW, Wang GJ, Alexoff D, Logan J, Muench L, Shea C, Telang F, Fowler JS, Wong C, Benveniste H, Tomasi D (2013) Acute alcohol intoxication decreases glucose metabolism but increases acetate uptake in the human brain. NeuroImage 64:277-283.

Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding YS, Logan J, Wong C, Miller EN (2001c) Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. The American journal of psychiatry 158:377-382.

Volkow ND, Chang L, Wang GJ, Fowler JS, Ding YS, Sedler M, Logan J, Franceschi D, Gatley J, Hitzemann R, Gifford A, Wong C, Pappas N (2001d) Low level of brain dopamine D2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex. The American journal of psychiatry 158:2015-2021.

Wagner GC, Ricaurte GA, Seiden LS, Schuster CR, Miller RJ, Westley J (1980) Long-lasting depletions of striatal dopamine and loss of dopamine uptake sites following repeated administration of methamphetamine. Brain Res 181:151-160.

Wang GJ, Volkow ND, Chang L, Miller E, Sedler M, Hitzemann R, Zhu W, Logan J, Ma Y, Fowler JS (2004) Partial recovery of brain metabolism in methamphetamine abusers after protracted abstinence. The American journal of psychiatry 161:242-248.

Wang X, Su B, Siedlak SL, Moreira PI, Fujioka H, Wang Y, Casadesus G, Zhu X (2008) Amyloid-beta overproduction causes abnormal mitochondrial dynamics via differential modulation of mitochondrial fission/fusion proteins. Proc Natl Acad Sci U S A 105:19318-19323.

Wang ZH, Ding MX, Chew-Cheng SB, Yun JP, Chew EC (1999) Bcl-2 and Bax proteins are nuclear matrix associated proteins. Anticancer Res 19:5445-5449.

Wawrzenczyk A, Nalecz KA, Nalecz MJ (1995) Effect of externally added carnitine on the synthesis of acetylcholine in rat cerebral cortex cells. Neurochem Int 26:635-641.

Weng SJ, Shiue CY, Huang WS, Cheng CY, Huang SY, Li IH, Tao CC, Chou TK, Liao MH, Chang YP, Ma KH (2012) PET imaging of serotonin transporters with 4-[(18)F]-ADAM in a Parkinsonian rat model. Cell transplantation.

Winter BK, Fiskum G, Gallo LL (1995) Effects of L-carnitine on serum triglyceride and cytokine levels in rat models of cachexia and septic shock. Br J Cancer 72:1173-1179.

Wise RA (2009) Roles for nigrostriatal--not just mesocorticolimbic--dopamine in reward and addiction. Trends in neurosciences 32:517-524.

Wu CW, Ping YH, Yen JC, Chang CY, Wang SF, Yeh CL, Chi CW, Lee HC (2007) Enhanced oxidative stress and aberrant mitochondrial biogenesis in human neuroblastoma SH-SY5Y cells during methamphetamine induced apoptosis. Toxicol Appl Pharmacol 220:243-251.

Wu X, Huang W, Prasad PD, Seth P, Rajan DP, Leibach FH, Chen J, Conway SJ, Ganapathy V (1999) Functional characteristics and tissue distribution pattern of organic cation transporter 2 (OCTN2), an organic cation/carnitine transporter. J Pharmacol Exp Ther 290:1482-1492.

Wyss V, Ganzit GP, Rienzi A (1990) Effects of L-carnitine administration on VO2max and the aerobic-anaerobic threshold in normoxia and acute hypoxia. Eur J Appl Physiol Occup Physiol 60:1-6.

Yagi S, Yoshikawa E, Futatsubashi M, Yokokura M, Yoshihara Y, Torizuka T, Ouchi Y (2010) Progression from unilateral to bilateral parkinsonism in early Parkinson disease: implication of mesocortical dopamine dysfunction by PET. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 51:1250-1257.

Yamaguti K, Kuratsune H, Watanabe Y, Takahashi M, Nakamoto I, Machii T, Jacobsson G, Onoe H, Matsumura K, Valind S, Langstrom B, Kitani T (1996) Acylcarnitine metabolism during fasting and after refeeding. Biochem Biophys Res Commun 225:740-746.

Yamamoto BK, Zhu W (1998) The effects of methamphetamine on the production of free radicals and oxidative stress. J Pharmacol Exp Ther 287:107-114.

Yudkoff M (1997) Brain metabolism of branched-chain amino acids. Glia 21:92-98.

Page 139: ÍDIA LEXANDRA DOS ANTOS CUNHA - Repositório … · lower ATP synthesis, reduced mitochondrial function, higher levels of intracellular ROS, decreased mitochondrial membrane potential

139

Zaitone SA, Abo-Elmatty DM, Shaalan AA (2011) Acetyl-L-carnitine and alpha-lipoic acid affect rotenone-induced damage in nigral dopaminergic neurons of rat brain, implication for Parkinson's disease therapy. Pharmacol Biochem Behav 100:347-360.

Zanelli SA, Solenski NJ, Rosenthal RE, Fiskum G (2005) Mechanisms of ischemic neuroprotection by acetyl-L-carnitine. Ann N Y Acad Sci 1053:153-161.

Zhang R, Zhang H, Zhang Z, Wang T, Niu J, Cui D, Xu S (2012) Neuroprotective effects of pre-treament with L-carnitine and acetyl-L-carnitine on ischemic injury in vivo and in vitro. Int J Mol Sci 13:2078-2090.

Zingarelli B, O'Connor M, Wong H, Salzman AL, Szabo C (1996) Peroxynitrite-mediated DNA strand breakage activates poly-adenosine diphosphate ribosyl synthetase and causes cellular energy depletion in macrophages stimulated with bacterial lipopolysaccharide. J Immunol 156:350-358.