brain 2012 aviles olmos brain aws009 (1)

Upload: rubens-andrade

Post on 14-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    1/11

    BRAINA JOURNAL OF NEUROLOGY

    REVIEW ARTICLE

    Parkinsons disease, insulin resistance and novelagents of neuroprotectionIciar Aviles-Olmos,1 Patricia Limousin,1 Andrew Lees2 and Thomas Foltynie1

    1 Sobell Department of Motor Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK

    2 Reta Lila Weston Institute, 1 Wakefield Street, London WC1N 1PJ, UK

    Correspondence to: Thomas Foltynie,

    Box 146, National Hospital for Neurology and Neurosurgery,

    Queen Square,

    London WC1N 3BG, UK

    E-mail: [email protected]

    Multiple avenues of research including epidemiology, molecular genetics and cell biology have identified links between

    Parkinsons disease and type 2 diabetes mellitus. Several recent discoveries have highlighted common cellular pathways that

    potentially relate neurodegenerative processes with abnormal mitochondrial function and abnormal glucose metabolism. This

    includes converging evidence identifying that peroxisome proliferator activated receptor gamma coactivator 1-a, a key regulator

    of enzymes involved in mitochondrial respiration and insulin resistance, is potentially pivotal in the pathogenesis of neurode-

    generation in Parkinsons disease. This evidence supports further study of these pathways, most importantly to identify neu-

    roprotective agents for Parkinsons disease, and/or establish more effective prevention or treatment for type 2 diabetes mellitus.

    In parallel with these advances, there are already randomized trials evaluating several established treatments for insulin resist-

    ance (pioglitazone and exenatide) as possible disease modifying drugs in Parkinsons disease, with only preliminary insights

    regarding their mechanisms of action in neurodegeneration, which may be effective in both disease processes through an action

    on mitochondrial function. Furthermore, parallels are also emerging between these same pathways and neurodegeneration

    associated with Alzheimers disease and Huntingtons disease. Our aim is to highlight this converging evidence and stimulate

    further hypothesis-testing studies specifically with reference to the potential development of novel neuroprotective agents in

    Parkinsons disease.

    Keywords: Parkinsons disease; type 2 diabetes; neuroprotection; Alzheimers disease

    Abbreviations: PGC1 = peroxisome proliferator activated receptor gamma coactivator 1 ; MPTP = 1-methyl 4-phenyl1,2,3,6-tetrahydropyridine; PINK1 = PTEN-induced putative kinase 1; PARIS = parkin interacting substrate

    IntroductionThe pathogenesis of Parkinsons disease is gradually beingelucidated through the combined efforts of epidemiologists, gen-

    eticists and molecular and cell biologists. Overlapping and inter-

    related pathways involving mitochondrial turnover (mitophagy and

    mitochondrial biogenesis), neuroinflammation and aggregation

    and disaggregation of toxic protein oligomers, all appear to be

    contributory. While these discoveries represent clear progress,

    from the patients perspective, there remains a greater urgencyto search for and test potential neuroprotective or even neuror-

    estorative treatments even prior to a definitive understanding of

    disease pathogenesis.

    Important steps in this neurodegenerative pathway have recently

    been discovered highlighting links between Parkinsons disease

    pathogenesis and the mechanisms underlying the development of

    doi:10.1093/brain/aws009 Brain 2012: Page 1 of 11 | 1

    Received August 18, 2011. Revised November 21, 2011. Accepted November 25, 2011.

    The Author (2012). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved.

    For Permissions, please email: [email protected]

    Brain Advance Access published February 17, 2012

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    2/11

    insulin resistance. This is particularly noteworthy since several agents

    used in the treatment of type 2 diabetes have been shown to be

    neuroprotective in animal models of Parkinsons disease and are

    now being evaluated in randomized controlled trials in patients with

    Parkinsons disease. This review provides a synthesis of the emerging

    links between Parkinsons diseasepathogenesis and insulin resistance

    to generate hypotheses that can be further confirmed or refuted, and

    to describe the basis for the investigation of anti-diabetes agents as

    possible neuroprotective agents in Parkinsons disease.

    Search strategyReferences for this review were identified through searches

    of PubMed (http://www.ncbi.nlm.nih.gov/pubmed/) with the

    search terms: Parkinsons disease, neurodegeneration, diabetes,

    insulin resistance, mitochondrial, PGC1, pioglitazone and exena-

    tide from 1990 to May 2011. Articles were also obtained through

    searches of the authors own files. Only papers published in

    English were reviewed. The final reference list was generated on

    the basis of relevance to the broad scope of this review.

    Links between Parkinsons disease andtype 2 diabetes mellitus

    Epidemiology

    Evidence from prospective epidemiological studies has identified type

    2 diabetes mellitus as an independent risk factor for multiple diseases

    of the nervous system such as diabetic neuropathy (Boulton et al.,

    2005), stroke (Tuomilehto et al., 1996; Hu et al., 2006) and more

    recently Alzheimers disease (Leibson et al., 1997; Ott et al., 1999;

    Peila et al., 2002). There are also reports of varying associations be-

    tween diabetes or abnormal glucose tolerance and sporadic forms ofParkinsons disease from both cross-sectional and cohort studies.

    Survey data reveal that diabetes is established in 830% of patients

    with Parkinsons disease, consistently in excess of the prevalence

    found in non-Parkinsons disease individuals (Chalmanov and

    Vurbanova, 1987; Pressley et al., 2003). This might be readily ex-

    plained by increased detection of hyperglycaemia through additional

    medical contact/urine/blood tests among individuals with

    Parkinsons disease regularly attending hospital appointments. The

    association is strengthened by reports suggesting that up to 5080%

    of patients with Parkinsons disease haveabnormal glucose tolerance

    when tested (although these figures are from non-contemporary

    papers and we are unaware of confirmatory data from more recent

    cohorts) (Barbeau etal., 1961; Elner and Kandel,1965; Lipman etal.,1974; Sandyk, 1993); however, in a series of 800 patients with

    Parkinsons disease, concurrent diabetes was indeed shown to accel-

    erate progression of both motor and cognitive symptoms (Schwab,

    1960). In view of the possible confounding effects of Parkinsons

    disease treatment, newly diagnosed, never-treated adults with

    Parkinsons disease have also been studied and been shown to have

    reduced insulin-mediated glucose uptake (Van Woert and Mueller,

    1971), inhibition of early insulin secretion and long-term hyperinsu-

    linaemia and hyperglycaemia after glucose loading (Boyd et al.,

    1971). This takes into account the effects of some drugs used to

    treat Parkinsons disease, such as levodopa, which induces both

    hyperglycaemia and hyperinsulinaemia, whereas others (including

    the ergot dopamine agonist bromocriptine) may increase insulin sen-

    sitivity (Van Woert and Mueller, 1971; Sirtori et al., 1972).

    Neuropathological studies of patients with Parkinsons disease

    have shown that insulin receptors are densely represented on

    the dopaminergic neurons of the substantia nigra pars compacta

    (Unger et al., 1991) and loss of insulin receptor immunoreactivity

    and messenger RNA in the substantia nigra pars compacta of pa-

    tients with Parkinsons disease coincides with loss of tyrosine

    hydroxylase messenger RNA (the rate-limiting enzyme in dopa-

    mine synthesis) (Moroo et al., 1994; Takahashi et al., 1996).

    Indeed abnormal glucose utilization has been specifically shown

    in the brains of patients with Parkinsons disease undergoing

    either magnetic resonance spectroscopy (Bowen et al., 1995) or

    fluorodeoxyglucose-PET (Hu et al., 2000) demonstrating increased

    lactate concentrations and glucose hypometabolism, supporting

    the hypothesis that Parkinsons disease is a systemic disorder char-

    acterized by a derangement of oxidative energy metabolism.

    Further evidence for a positive association between Parkinsons

    disease and type 2 diabetes mellitus has been obtained from pro-spective cohort studies. A statistically significant direct association

    between triceps skinfold thickness and the risk of Parkinsons dis-

    ease has been found in the Honolulu Heart Program (Abbott

    et al., 2002) and both excess weight (Hu et al., 2006) and type

    2 diabetes mellitus itself (Hu et al., 2007) were associated with an

    increased risk of Parkinsons disease in a population-based pro-

    spective cohort of Finnish males and females. This association

    was independent of the known modifying factors such as smoking

    status, coffee and alcohol consumption and body weight, tempt-

    ing speculation regarding common pathways underlying the de-

    velopment of these conditions. Nevertheless, a recent cohort study

    could not replicate an association between either type 2 diabetes

    mellitus or obesity and Parkinsons disease risk although the au-thors acknowledge that diagnosis of type 2 diabetes mellitus was

    entirely based on self-report (Palacios et al., 2011).

    Hyperglycaemia and dopamine

    Any link between Parkinsons disease and type 2 diabetes mellitus

    may relate to abnormalities in a common pathway or indirectly as

    a consequence of chronic hyperglycaemia or hyperinsulinaemia.

    Animal and in vitro studies have suggested a role for insulin in

    the regulation of brain dopaminergic activity featuring a reciprocal

    regulation between the two chemicals (Craft and Watson, 2004).

    In the rat, elevation of glucose concentrations in the blood, to

    levels equivalent to those produced by a meal or stress, suppresses

    the firing of dopamine-containing neurons located within the sub-stantia nigra and prevents or reverses the increase in discharge

    rates of dopaminergic cells normally elicited by the dopamine-

    receptor antagonist haloperidol (Saller and Chiodo, 1980).

    Administration of glucose to rats has also been shown to produce

    a significant decrease of dopamine turnover in both striatum

    and olfactory tubercle (Montefusco et al., 1983). Furthermore,

    hyperglycaemia related to hypoinsulinaemia resulting from

    streptozotocin-induced diabetes (which leads to toxicity to b islet

    cells and is a model of type 1 diabetes mellitus) has been shown to

    decrease basal dopamine concentrations and amphetamine-

    2 | Brain 2012: Page 2 of 11 I. Aviles-Olmos et al.

    http://www.ncbi.nlm.nih.gov/pubmed/http://www.ncbi.nlm.nih.gov/pubmed/
  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    3/11

    induced dopamine overflow in the mesolimbic cortex (Murzi et al.,

    1996).

    Chronic hyperglycaemia leads to oxidative stress and the pro-

    duction of reactive oxygen species, which has been implicated in

    the ongoing b cell death in type 2 diabetes mellitus (Evans et al.,

    2002). While production of reactive oxygen species may also be a

    mechanism underlying dopaminergic cell loss in hyperglycaemic

    animals, this remains speculative, and a further possibility emerges

    from work demonstrating that in both insulin-deficient rats and

    insulin-resistant mice, diabetes impairs hippocampus-dependent

    memory, synaptic plasticity and adult neurogenesis (Stranahan

    et al., 2008, 2010). However, given that any epidemiological

    association between Parkinsons disease and dopamine appears

    restricted to type 2 diabetes mellitus, it is likely that chronic hyper-

    glycaemia per se, is only a minor risk factor for Parkinsons disease

    in humans.

    Insulin resistance and mitochondrialdysfunction

    The development of insulin resistance in humans is closely corre-

    lated with immune cell infiltration and inflammation, with clear

    links between the development of obesity, type 2 diabetes mellitus

    and cardiovascular disease (Hotamisligil, 2006). Unambiguous links

    have also been established between exercise, obesity, insulin

    resistance and levels of interleukin-6 (Kern et al., 2001).

    Evidence for mitochondrial dysfunction in the development of in-

    sulin resistance has been obtained through measuring rates of

    in vivo mitochondrial phosphorylation using proton 1(H) magnetic

    resonance spectroscopy in the relatives of patients with type 2

    diabetes mellitus, finding that rates of mitochondrial ATP produc-

    tion are reduced by 30% in the muscle of lean, pre-diabetic insulin

    resistant subjects (Petersen et al., 2004). In addition, there aremany specific examples of insulin resistance occurring due to

    mitochondrial mutations; it has been estimated that $1.5% of

    type 2 diabetes mellitus is attributable to the mitochondrial

    A3243G mutation [the cause of mitochondrial encephalopathy,

    lactic acidosis and stroke-like episodes (MELAS; Gerbitz et al.,

    1995)].

    The more widespread development of insulin resistance has also

    been proposed to be mediated through mitochondrial dysfunction

    (Stark and Roden, 2007) at least to a partial extent through

    muscle expression of transcriptional regulators including PGC1,

    an important regulator of enzymes involved in mitochondrial

    respiration. Expression of PGC1 is rapidly induced following

    even a single bout of exercise, which then reverts to baselineafter cessation of exercise to enable fine control of the energy

    demands of skeletal muscle (Handschin and Spiegelman, 2008).

    Individuals that undergo regular exercise have chronically elevated

    levels of PGC1, in association with a switch in muscle fibre type

    characterized by increased mitochondrial density and function

    (Lin et al., 2002).

    Direct exploration of patterns of gene expression associated

    with insulin resistance has been studied in skeletal muscle in

    patients with type 2 diabetes mellitus as well as non-diabetic in-

    dividuals with and without a family history of type 2 diabetes

    mellitus. The earliest sign of insulin resistance was shown to be

    reduction in expression of PGC1 and the mitochondrial gene

    nuclear respiratory factor 1 (NRF1) (Patti et al., 2003) . I n a

    whole genome methylation analysis of skeletal muscle from type

    2 diabetes mellitus and control subjects, hypermethylation of

    PGC1 was found in association with reduced PGC1 messenger

    RNA and reduced mitochondrial DNA levels in subjects with type 2

    diabetes mellitus. Hypermethylation of PGC1 can be induced by

    dietary factors such as fatty acids, as well as cytokines such as

    tumour necrosis factor- (TNF-), suggesting a potential mechan-

    ism underlying the geneenvironment interaction in type 2 dia-

    betes mellitus risk (Barres et al., 2009).

    Parkinsons disease and mitochondrialdysfunction

    The earliest link between Parkinsons disease and mitochondrial

    dysfunction followed the observation that individuals injected

    with heroin contaminated with MPTP (1-methyl 4-phenyl

    1,2,3,6-tetrahydropyridine) acutely developed parkinsonism

    (Langston et al., 1983). Both MPTP and another environmental

    toxin, rotenone, were subsequently shown to cause degeneration

    of dopaminergic neurons in animal models by selective inhibition

    of complex I (Gerlach et al., 1991) suggesting that in humans,

    complex I impairment may itself be sufficient to cause the disease.

    Complex I (NADH CoQ dehydrogenase) is the first enzyme of the

    mitochondrial respiratory chain, playing a crucial role in ATP gen-

    eration. Complex I dysfunction decreases ATP production, gener-

    ates free radicals and sensitizes cells to the pro-apoptotic protein

    Bax thus leading to apoptosis (Perier et al., 2005). A direct relation

    between mitochondrial dysfunction and sporadic Parkinsons dis-

    ease has been demonstrated in post-mortem tissue, revealing

    complex I deficiency (Schapira et al., 1989; Mann et al., 1994)and damage (Keeney et al., 2006) in the substantia nigra of pa-

    tients with Parkinsons disease. However, it is likely that neurode-

    generation provoked by complex I inhibitors alone is rare.

    Complex I toxins may, however, contribute to the pathogenesis

    of Parkinsons disease acting in tandem with other environmental

    influences and genetic causes (Schapira, 2010). Although also rare,

    there are numerous reports of parkinsonism occurring in associ-

    ation with mitochondrial mutations including the G11778A muta-

    tion associated with Lebers hereditary optic neuropathy (Simon

    et al., 1999), polymerase- mutations (Luoma et al., 2007) and

    more commonly in association with somatic mitochondrial dele-

    tions (Bender et al., 2006). A common mitochondrial haplotype

    has also been associated with sporadic Parkinsons disease(Pyle et al., 2005).

    Converging evidence suggests that both environmental and

    genetic factors can interfere with normal processes of the removal

    of damaged or dysfunctional mitochondria known as mitophagy.

    Healthy mitochondrial turnover through mitophagy and mitochon-

    drial biogenesis as well as ongoing processes of fission, fusion and

    mitochondrial budding are all essential to maintain normal cellular

    bioenergetic function. This relies on intact mitochondrial DNA,

    which is particularly vulnerable to damage by free radicals due

    to its lack of a histone coat and limited facilities for repair.

    Parkinsons disease and insulin resistance Brain 2012: Page 3 of 11 | 3

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    4/11

    Furthermore mitochondrial DNA mutations, whether induced by

    oxidative stress or ageing, have a further effect on complex I and

    respiratory chain function leading to increased damage again

    through the production of reactive oxygen species (Cooper

    et al., 1992). Normal human substantia nigra and striatum exhibit

    the greatest free radical-mediated mitochondrial damage with age

    (Kraytsberg et al., 2006). Other cellular pathways essential for

    normal mitochondrial turnover have been identified directly as a

    result of the identification of specific gene mutations causing

    Mendelian forms of Parkinsons disease (Schapira, 2008).

    Parkin mutations cause autosomal recessive juvenile parkinson-

    ism. Intracellular localization studies have reported the association

    of parkin and mitochondria (Stichel et al., 2000). The function of

    parkin is not completely clear but the protein has E3 ligase activity,

    and is thought to monitor the quality of mitochondria and trigger

    mitophagy of dysfunctional mitochondria (Rakovic et al., 2010),

    by triggering the ubiquitinproteasome system (Chan et al.,

    2011). Parkin knockout mice have demonstrated alterations in

    abundance and/or modification of a number of proteins involved

    in mitochondrial function or oxidative stress, with reductions in

    several subunits of complexes I and IV, and functional assaysshowing reductions in respiratory capacity of striatal mitochondria

    isolated from parkin/ mice. Furthermore, these mice show a

    delayed rate of weight gain, suggesting broader metabolic

    abnormalities (Palacino et al., 2004). Mitochondrial function has

    also been shown to be decreased (complex I and IV activities) in

    peripheral blood from patients with parkin mutations (Muftuoglu

    et al., 2004).

    PINK1 (PTEN-induced putative kinase 1) mutations also cause

    autosomal recessive juvenile parkinsonism. The gene encoding

    PINK1 encodes a 63-kDa protein with an 8-kDa mitochondrial

    targeting sequence. In health, this protein is imported intact into

    the mitochondria, where it has been suggested that PINK 1 re-

    cruits parkin from the cytoplasm to the mitochondria to initiate theprocess of mitophagy (Vives-Bauza and Przedborski, 2011).

    DJ-1 is a 23-kDa protein that is expressed in peripheral tissues

    and parts of the brain, including the hippocampus, cerebellum,

    olfactory bulb, striatum, substantia nigra pars compacta and sub-

    stantia nigra pars reticulata, both in cells bodies and dendrites,

    localized to the mitochondrial matrix and intermembrane space

    (Zhang et al., 2005). The deletion or silencing of DJ-1 causes

    parkinsonism possibly by sensitising cells to oxidative stress,

    while over-expression of DJ-1 protects cells implying a protective

    role for the protein (Yokota et al., 2003). Substantia nigra neurons

    from DJ-1 knockout mice have increased sensitivity to MPTP

    and oxidative stress (Kim et al., 2005), while cells derived from

    patients with DJ-1 mutations have abnormal mitochondrial morph-ology (Irrcheret al., 2010). DJ-1 has been shown to associate with

    the mitochondrial Bcl-XL, an anti-apoptotic protein (Ren et al.,

    2011).

    -Synuclein (SNCA) mutations can cause autosomal dominant

    Parkinsons disease, and -synuclein is the main component of the

    pathological feature of Parkinsons diseasethe Lewy body. The

    pathological features of Parkinsons disease, can be replicated

    by simply over-expressing SNCA in transgenic flies (Feany and

    Bender, 2000) suggesting that one mechanism by which neurode-

    generation occurs is through a toxic gain-of-function. The exact

    function of -synuclein remains unknown, and the relationship

    between SNCA oligomers and aggregates, their degradation by

    the ubiquitinproteasome and lysosomal systems and neuronal

    toxicity requires further work (Wong and Cuervo, 2010). There

    are nevertheless indications that SNCA affects various mitochon-

    drial pathways (Poon et al., 2005). SNCA co-localizes with cyto-

    chrome c forming hetero-oligomers, which can prevent apoptosis,

    but in the process forms complexes with prolonged peroxidase

    activity that induces protracted oxidative stress (Bayir et al.,

    2009). It has also been shown that phosphorylated SNCA (the

    dominant form in Parkinsons disease) influences the normal pro-

    teinprotein interactions including the pull down of protein com-

    plexes involved in mitochondrial electron transport (McFarland

    et al., 2008), while over-expression of SNCA leads to the protein

    entering mitochondria and interfering with mitochondrial function

    (Devi et al., 2008; Chinta et al., 2010).

    Leucine-rich repeat kinase (LRRK-2) mutations can also cause

    autosomal dominant Parkinsons disease. This protein has GTPase

    and kinase domains and study of its interaction with SNCA is on-

    going (Greggio et al., 2011). Whether LRRK-2 plays a critical role

    in determining the phosphorylation status of SNCA remains to bedetermined, however, it has recently been demonstrated that

    in the transgenic G2019S mutant LRRK2 mouse, there is age-

    dependent degeneration of dopamine nigrostriatal neurons to-

    gether with damaged mitochondria and an increase in mitophagy

    (Ramonet et al., 2011).

    Glucocerebrosidase (GBA) mutations cause Gauchers disease

    and have been identified as a risk factor for Parkinsons disease

    even in the heterozygous state (Sidransky et al., 2009). There has

    been shown to be a bidirectional link such that SNCA inhibits the

    lysosomal activity of GBA and functional loss of GBA leads to

    accumulation of SNCA (Mazzulli et al., 2011), potentially explain-

    ing the risk of Parkinsons disease through a positive feedback

    loop. Other potential mechanisms also include lipid accumulationand impaired mitophagy or mitochondrial trafficking (Westbroek

    et al., 2011).

    In Parkinsons disease there is therefore ample evidence of mito-

    chondrial dysfunction that includes complex I inhibition, oxidative

    stress, PINK1 and DJ-1 dysfunction, and an interaction between

    parkin and PINK1 that influences mitophagy and mitochondrial

    biogenesis. Complex I inhibition initiated by any of a range of

    environmental toxins will increase free radical generation, and

    thus initiate a vicious cycle of events that further impairs mito-

    chondrial function and may enhance any underlying genetic

    defects and further impair neuronal activity. Neuroinflammation

    undoubtedly leads to further mitochondrial stress through the pro-

    duction of reactive oxygen species as well as through the activa-tion of microglia and subsequent release of pro-inflammatory

    cytokines such as nitric oxide and TNF- (Whitton, 2007; Tansey

    et al., 2008). This can be readily detected through the identifica-

    tion of activated microglia on imaging (Gerhard et al., 2006) and

    neuropathology (McGeer et al., 1988). The role of a neuroinflam-

    matory process in Parkinsons disease is further supported by the

    association between the human leukocyte antigen locus and

    Parkinsons disease risk in a meta-analysis of genome wide studies

    (Nalls et al., 2011) and the reported beneficial effects of

    anti-inflammatory agents on risk of Parkinsons disease (Wahner

    4 | Brain 2012: Page 4 of 11 I. Aviles-Olmos et al.

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    5/11

    et al., 2007). It is likely that multiple pathways remain relevant in

    Parkinsons disease pathogenesis with interrelated aspects that

    impact on mitochondrial function.

    Converging evidence implicatingPGC1a

    Converging evidence suggests that cellular pathways leading toeither insulin resistance or neurodegeneration involve mitochon-

    drial mechanisms. It is well known that mutations in mitochondrial

    DNA can lead to a wide variety of phenotypes that commonly

    involve neurodegeneration and diabetes (Hara et al., 1994;

    Finsterer et al., 2008). However, these mutations do not account

    for a significant proportion of cases of sporadic Parkinsons dis-

    ease. Normal mitochondrial biogenesis, respiration and metabolism

    of reactive oxygen species requires intact expression of both

    nuclear and mitochondrial encoded genomes now recognized as

    being regulated by PGC1 (Lin et al., 2005; Finck and Kelly, 2006;

    St-Pierre et al., 2006). It has previously been shown that PGC1

    has a powerful suppressive effect on reactive oxygen species pro-

    duction, in parallel to its effects in elevating mitochondrial respir-

    ation. This occurs through the PGC1-mediated expression of

    genes involved in reactive oxygen species detoxification, as well

    as PGC1 expression that is rapidly induced by these proteins

    following a single bout of endurance exercise in vivo (Handschin

    and Spiegelman, 2008). Insulin resistant patients show reduced

    expression of PGC1 and the mitochondrial encoded gene

    COX1 (Heilbronn et al., 2007), while reduction in PGC1-

    responsive genes has been shown among patients with type 2

    diabetes mellitus and their asymptomatic relatives compared with

    healthy controls (Petersen et al., 2004). Indeed polymorphisms in

    PGC1 have been associated with an increased risk for type 2

    diabetes mellitus in diverse populations (Ek et al., 2001; Haraet al., 2002; Bhat et al., 2007).

    PGC1 has also been implicated as having a major role in

    Parkinsons disease pathogenesis. Meta-analysis of gene expres-

    sion data using microarrays has utilized post-mortem brain hom-

    ogenates to look at gene expression in the substantia nigra pars

    compacta of patients with confirmed SNCA-positive Lewy body

    Parkinsons disease. Robust three tiered analysis from separate

    genome wide datasets have indicated that gene sets involved

    in mitochondrial electron transport, mitochondrial biogenesis,

    glucose utilization and glucose sensing were strongly associated

    with Parkinsons disease. Included amongst these gene sets were

    10 PGC1 responsive genes. Furthermore it was shown that

    over-expression of PGC1 was able to protect dopaminecell loss induced by the mitochondrial toxin rotenone (Zheng

    et al., 2010).

    In parallel with these discoveries was the identification of a

    zinc-finger protein, parkin interacting substrate (PARIS), which is

    up-regulated 3-fold in the nigra of patients with not only

    parkin-related parkinsonism but also sporadic Parkinsons disease,

    and is both necessary and sufficient for the neurodegeneration

    associated with parkin animal models (Shin et al., 2011). The

    same group further identified that PARIS represses the expression

    of PGC1 and PGC1 target genes playing an important role in

    mitochondrial function including NRF1, and the oxidative

    phosphorylation regulator ATP5b. The site of interaction between

    PARIS and PGC1 is a sequence that is involved in the regulation

    of transcripts involved in insulin responsiveness and energy metab-

    olism (Mounier and Posner, 2006). Although other pathways are

    undoubtedly also relevant, it is clear that parkin, PARIS, PGC1

    and NRF1 contribute to the pathogenesis of Parkinsons disease.

    Loss of expression of PGC1 controlled genes may therefore be a

    key link between abnormal mitochondrial function, abnormal glu-

    cose utilization and Parkinsons disease. Hypermethylation of

    PGC1 during life may follow either genetic or environmental in-

    fluences that promote accumulation of free fatty acids, TNF- and

    ceramides (Summers and Nelson, 2005; Staiger et al., 2006; Barres

    et al., 2009), which might then lead to decompensation of mito-

    chondrial bioenergetics and the onset of Parkinsons disease

    (Figure 1).

    Tissue specificity of mitochondrialdysfunction: parallels between insulin

    resistance, Parkinsons disease andother neurodegenerative diseases

    There has been considerable speculation regarding the (initially)

    selective degeneration of dopaminergic neurons in Parkinsons dis-

    ease. Dopaminergic neurons in the substantia nigra pars compacta

    have an incredibly complex structure in terms of axon length and

    number of synapses (4100 000). Mathematical modelling has

    shown that neuronal energetic demand rises exponentially with

    axon structure and arbour complexity. This potentially makes

    dopaminergic neurons of the substantia nigra pars compacta

    more sensitive to energetic stress than other types of dopamine

    or indeed non-dopaminergic neurons (Matsuda et al., 2009; Moss

    and Bolam, 2010).There is considerable evidence from epidemiology, neuropath-

    ology and functional neuroimaging that also implicates diabetes as

    a risk factor for cognitive impairment and Alzheimers disease

    (Janson et al, 2004; Luchsinger et al., 2007; Toro et al., 2009).

    In a study of animal models of double-mutant Alzheimer and dia-

    betic transgenic mice, the onset of diabetes has been shown to

    exacerbate Alzheimers disease-like cognitive dysfunction (Takeda

    et al., 2010). Excessive energy intake appears to affect cognitive

    function adversely though oxidative stress, inflammation and im-

    paired cellular stress responses (Pistell et al., 2010; Kapogiannis

    and Mattson, 2011), mediated via reduced expression and/or ac-

    tivity of mitochondrial proteins and oxidative genomic damage

    (Bishop et al., 2010). In animal models in which neurotoxicity ismediated by oxidative stress, dietary energy restriction has been

    shown to protect neurons and synapses (Guo et al., 2000), while

    exercise has been shown to enhance hippocampal neurogenesis

    and reduce glucocorticoid levels (Kannangara et al., 2010).

    Ageing and chronic hyperinsulinaemia both upregulate genes

    for inflammatory/immune pathways and downregulate insulin sig-

    nalling genes, blocking glucose utilization and decreasing mito-

    chondrial function in hippocampal neurons (Blalock et al., 2010).

    This is reflected in the reduction in the activity of several mito-

    chondrial enzymes (e.g. pyruvate and isocitrate dehydrogenases,

    Parkinsons disease and insulin resistance Brain 2012: Page 5 of 11 | 5

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    6/11

    -ketoglutarate dehydrogenase complex) in brain tissue samples

    from patients with Alzheimers disease (Bubber et al., 2005).

    Abnormal distribution of mitochondria in fibroblasts taken from

    subjects with Alzheimers disease has been proposed as evidence

    that mitochondrial fission/fusion processes may have relevance in

    Alzheimers pathogenesis (Wang et al., 2008). Furthermore, there

    are data to show an association between reduced PGC1 levels

    and Alzheimer pathology in post-mortem human brains, and that

    PGC1 over-expression can reduce hyperglycaemia-mediated

    b amyloidogenesis (Qin et al., 2009).

    In parallel with the complexity of nigro-striatal neurons, hippo-

    campal pyramidal neurons have the highest energy requirements

    of any neurons in the brain (LaManna and Harik, 1985) and this

    may also contribute to their vulnerability to neurotoxicity mediatedvia mitochondrial dysfunction in Alzheimers disease. Analogous

    with Parkinsons disease pathogenesis, it is likely that a combin-

    ation of factors including amyloid-b oligomer accumulation, oxi-

    dative stress as well as a deficit in neurotrophic factor signalling all

    play a role in mitochondrial dysfunction in vulnerable neurons

    producing mild cognitive impairment and Alzheimers disease

    (Mattson et al., 2008). The association of molecular alterations

    in Alzheimers disease with perturbed neuronal energy metabolism

    and the impact of energy intake and expenditure on cognition and

    ageing suggest an important link between Alzheimers disease

    pathogenesis and brain energy metabolism that may be amenable

    to pharmacological interventions.

    Other neurodegenerative diseases also share links with type 2

    diabetes mellitus. Patients with Huntingtons disease develop type

    2 diabetes mellitus about seven times more often than matched

    healthy controls (Podolsky, 1972), and this is a consistent feature

    in the mouse models of Huntingtons disease (Hurlbert et al.,

    1999), with ensuing demonstrations that the mutant huntingtin

    protein has a direct effect on mitochondrial function and traffick-

    ing (Orr et al., 2008) and inhibits expression of PGC1 (Cui et al.,

    2006). In addition, type 2 diabetes mellitus is a common feature of

    Friedreichs ataxia, aceruloplasminaemia and ataxia telangiectasia,

    as well as a range of more esoteric disorders featuring neurode-

    generation, reviewed in Ristow et al. (2004).

    Therapeutic implications

    The converging evidence implicating the PARIS/PGC1 pathway

    in the neurodegenerative process associated with Parkinsons dis-

    ease highlights the need for further study of the therapeutic

    effects of PARIS inhibition, and/or PGC1 stimulation as potential

    neuroprotective treatments. High-throughput screens can identify

    potential compounds for in vitro and in vivo testing ahead of

    human trials. Particularly exciting is the realization that some of

    Figure 1 Schematic overview of emerging pathways linking insulin resistance and neurodegeneration. FFAS = free fatty acids;HLA = human leucocyte antigen; IL-6 = interleukin 6; LRRK-2 = leucine-rich repeat kinase 2; NO = nitric oxide; PGCI = ppar receptor

    coactivator; ROS = reactive oxygen species; UPS = ubiquitin proteasome system; SNCA = synuclein.

    6 | Brain 2012: Page 6 of 11 I. Aviles-Olmos et al.

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    7/11

    the existing licensed treatments for type 2 diabetes mellitus po-

    tentially have beneficial effects on related pathways of mitochon-

    drial function, directly or indirectly through an influence on PGC1

    activity, and trials have already been initiated to evaluate effects

    on patients with Parkinsons disease and/or other neurodegenera-

    tive diseases.

    Pioglitazone

    Pioglitazone is a licensed treatment for patients with type 2 dia-

    betes mellitus, and reduces insulin resistance via its action on the

    nuclear receptor peroxisome proliferator-activated receptor

    gamma (PPAR-). It modulates the transcription of genes involved

    in insulin sensitivity. Of major interest is the recent observation

    that pioglitazone and other active thiazolidinedione compounds

    bind to the outer mitochondrial membrane protein (mitoNEET)

    with an affinity comparable to its binding to PPAR- and there

    has been a suggestion that many of the clinical effects of

    pioglitazone are mediated by binding to mitoNEET (Colca et al.,

    2004; Paddock et al., 2007; Wiley et al., 2007). MitoNEET plays a

    key role in electron transport and oxidative phosphorylation, and

    pioglitazone binding to MitoNeet has been shown to have positiveregulatory effects on complex I activity in neuronal cells (Ghosh

    et al., 2007).

    Pioglitazone has been shown to be protective against neurode-

    generation in MPTP mouse models of Parkinsons disease (Breidert

    et al., 2002; Dehmer et al., 2004). The neuroprotective properties

    of pioglitazone have been suggested to be mediated via a sequen-

    tial action through PPAR activation, inducible nitric oxide synthase

    induction and nitric oxide-mediated toxicity (Dehmer et al., 2004).

    Pioglitazone has also been shown to be neuroprotective against

    the lipopolysaccharide model of Parkinsons disease through

    reduction of microglial activation and reduction of oxidative

    stress, enabling restoration of mitochondrial function (Hunter

    et al., 2007).Conflicting data have, however, suggested that the mechanism

    of action of pioglitazone in protection against MPTP induced

    neurodegeneration is solely mediated through the inhibition of

    monamine oxidase B (MAO-B), which prevents metabolism of

    MPTP to the toxic MPP+ (Quinn et al., 2008). In view of this

    uncertainty, but responding to the pressing need to evaluate this

    agent in patients with Parkinsons disease, a randomized trial has

    been initiated to explore possible neuroprotective effects of

    pioglitazone among patients already on MAO-B inhibitors

    (Clinical Trials.gov Identifier NCT01280123). Pioglitazone is also

    being trialled as a treatment for Alzheimers disease (Clinical

    Trials Identifier NCT00982202) and Friedreichs ataxia (Clinical

    Trials Identifier NCT00811681).

    Exenatide

    Glucagon-like peptide 1 (GLP-1) is a naturally occurring hormone

    that has an important role in insulin and glucose homeostasis but

    has a circulating half-life of only 12 min. Exenatide is a synthetic

    agonist for the GLP-1 receptor and has been granted a license for

    the treatment of type 2 diabetes with confirmed beneficial effects

    on glucose control, thought to be mediated by b-cell proliferation,

    increased insulin production, decreased gluconeogenesis and

    weight loss that follows chronic GLP-1 receptor stimulation in

    the gastrointestinal tract (Buse et al., 2004; DeFronzo et al.,

    2005; Kendall et al., 2005; Drucker et al., 2008). GLP-1 receptors

    are also distributed throughout the brain and stimulation of central

    receptors in the hypothalamus is responsible for early satiety.

    Further effects from central GLP-1 stimulation are as yet unclear,

    but neurotrophic and neuroprotective properties have been iden-

    tified in vitro (Perry et al., 2002). Exenatide has been evaluated as

    a neuroprotective agent in multiple animal models of Parkinsons

    disease (Bertilsson et al., 2008; Harkavyi et al., 2008; Kim et al.,

    2009; Li et al., 2009), demonstrating consistent benefits but again

    with inconsistent conclusions regarding mechanism of action

    whether anti-inflammatory (Harkavyi et al., 2008; Kim Chung

    et al., 2009; Kim et al., 2009) or related to stimulation of neuro-

    genesis (Bertilsson et al., 2008; Belsham et al., 2009; Li et al.,

    2010). More recently, exenatide has been shown to protect

    b islet cells from apoptosis, prevent damage to mitochondrial

    DNA encoded genes and stimulate mitochondrial biogenesis

    (Fan et al., 2010).

    Despite this mechanistic uncertainty (and even encouraged by

    the potential multiple effects on cell biology) and based on the

    encouraging data from the animal Parkinsons disease models, andthe excellent safety profile in patients with type 2 diabetes melli-

    tus, recruitment has been completed of patients to a randomized

    controlled trial at the UCL Institute of Neurology in Queen Square,

    London evaluating the effects of exenatide on Parkinsons disease

    (Clinical trials.gov Identifier NCT01174810).

    Scientists and physicians interested in Alzheimers disease are

    also studying the GLP-1 receptor agonists. Exenatide has been

    shown in animals to promote neuronal activity, enhance synaptic

    plasticity and cognitive performance (Perry and Greig, 2004).

    A randomized clinical trial to assess the safety and efficacy of

    exenatide treatment in participants with early Alzheimers disease

    is currently recruiting participants at NIA (Clinical trials.gov

    Identifier NCT01255163).

    ConclusionsA great deal of research has informed on the precise relationships

    between exercise, obesity, insulin resistance and cardiovascular

    risk. Links between type 2 diabetes mellitus and widely differing

    diseases are the subject of increasing interest, aside from the cur-

    rent hypothesis regarding Parkinsons disease, including possible

    relationships between diabetes and cancer (Giovannucci et al.,

    2010). Relationships between insulin, ageing and neurodegenera-

    tion have been considered in relation to toxic protein aggregation

    and the insulin-like growth factor-1 (IGF-1) signalling pathway(Cohen and Dillin, 2008). Alongside the undoubted importance

    of proteo-toxicity in neurodegeneration, the weight of converging

    evidence is highlighting the role of impaired mitochondrial func-

    tion in both Parkinsons disease and diabetes mellitus, mediated

    through PGC1. Therapeutic agents licensed for the treatment of

    type 2 diabetes mellitus and potentially having an impact on this

    pathway are already being evaluated in randomized controlled

    trials of patients with Parkinsons disease. Further identification

    of inhibitors and stimulators of the PGC1 pathway in neuronal

    cells will undoubtedly follow.

    Parkinsons disease and insulin resistance Brain 2012: Page 7 of 11 | 7

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    8/11

    FundingParkinsons Appeal, the Cure Parkinsons Trust and Parkinsons UK

    to T.F. None of these funding institutions had any influence or

    involvement in the writing of this article. The authors research

    into exenatide is being funded in its entirety by the Cure

    Parkinsons Trust.

    ReferencesAbbott RD, Ross GW, White LR, Nelson JS, Masaki KH, Tanner CM,

    et al. Midlife adiposity and the future risk of Parkinsons disease.

    Neurology 2002; 59: 10517.

    Armanini MP, Hutchins C, Stein BA, Sapolsky RM. Glucocorticoid endan-

    germent of hippocampal neurons is NMDA-receptor dependent. Brain

    Res 1990; 532: 712.

    Barbeau A, Giguere R, Hardy J. [Clinical experience with tolbutamide in

    Parkinsons disease]. Union Med Can 1961; 90: 14751.

    Barres R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, et al. Non-CpG

    methylation of the PGC-1a promoter through DNMT3B controls mito-

    chondrial density. Cell Metabolism 2009; 10: 18998.

    Bayir H, Kapralov AA, Jiang J, Huang Z, Tyurina YY, Tyurin VA, et al.

    Peroxidase mechanism of lipid-dependent cross-linking of synuclein

    with cytochrome C: protection against apoptosis versus delayed oxi-

    dative stress in Parkinson disease. J Biol Chem 2009; 284: 1595169.

    Belsham DD, Fick LJ, Dalvi PS, Centeno ML, Chalmers JA, Lee PK, et al.

    Ciliary neurotrophic factor recruitment of glucagon-like peptide-1

    mediates neurogenesis, allowing immortalization of adult murine

    hypothalamic neurons. FASEB J 2009; 23: 425665.

    Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK, Perry RH,

    et al. High levels of mitochondrial DNA deletions in substantia nigra

    neurons in aging and Parkinson disease. Nat Genet 2006; 38: 5157.

    Bertilsson G, Patrone C, Zachrisson O, Andersson A, Dannaeus K,

    Heidrich J, et al. Peptide hormone exendin-4 stimulates subventricular

    zone neurogenesis in the adult rodent brain and induces recovery in an

    animal model of Parkinsons disease. J Neurosci Res 2008; 86: 32638.

    Bhat A, Koul A, Rai E, Sharma S, Dhar MK, Bamezai RN. PGC-1alphaThr394Thr and Gly482Ser variants are significantly associated with

    T2DM in two North Indian populations: a replicate case-control

    study. Hum Genet 2007; 121: 60914.

    Bishop NA, Lu T, Yankner BA. Neural mechanisms of ageing and cogni-

    tive decline. Nature 2010; 464: 52935.

    Blalock EM, Grondin R, Chen KC, Thibault O, Thibault V, Pandya JD,

    et al. Aging-related gene expression in hippocampus proper compared

    with dentate gyrus is selectively associated with metabolic syndrome

    variables in rhesus monkeys. J Neurosci 2010; 30: 605871.

    Boulton AJ, Vinik AI, Arezzo JC, Bril V, Feldman EL, Freeman R, et al.

    Diabetic neuropathies: a statement by the American Diabetes

    Association. Diabetes Care 2005; 28: 95662.

    Bowen BC, Block RE, Sanchez-Ramos J, Pattany PM, Lampman DA,

    Murdoch JB, et al. Proton MR spectroscopy of the brain in 14 patients

    with Parkinson disease. AJNR Am J Neuroradiol;16: 618.Boyd AE 3rd, Lebovitz HE, Feldman JM. Endocrine function and glucose

    metabolism in patients with Parkinsons disease and their alternation

    by L-Dopa. J Clin Endocrinol Metab 1971; 33: 82937.

    Breidert T, Callebert J, Heneka MT, Landreth G, Launay JM, Hirsch EC.

    Protective action of the peroxisome proliferator-activated

    receptor-gamma agonist pioglitazone in a mouse model of

    Parkinsons disease. J Neurochem 2002; 82: 61524.

    Bubber P, Haroutunian V, Fisch G, Blass JP, Gibson GE. Mitochondrial

    abnormalities in Alzheimer brain: mechanistic implications. Ann Neurol

    2005; 57: 695703.

    Buse JB, Henry RR, Han J, Kim DD, Fineman MS, Baron AD. Effects of

    exenatide (exendin-4) on glycemic control over 30 weeks in

    sulfonylurea-treated patients with type 2 diabetes. Diabetes Care

    2004; 27: 262835.

    Chalmanov V, Vurbanova M. [Diabetes mellitus in parkinsonism

    patients]. Vutr Boles 1987; 26: 6873.

    Chan NC, Salazar AM, Pham AH, Sweredoski MJ, Kolawa NJ,

    Graham RL, et al. Broad activation of the ubiquitin-proteasome

    system by Parkin is critical for mitophagy. Hum Mol Genet 2011;

    20: 172637.

    Chinta SJ, Mallajosyula JK, Rane A, Andersen JK. Mitochondrial

    alpha-synuclein accumulation impairs complex function in dopamin-ergic neurons and results in increased mitophagy in vivo. Neurosci

    Lett 2010; 486: 2359.

    Cohen E, Dillin A. The insulin paradox: aging, proteotoxicity and neuro-

    degeneration. Nat Rev Neurosci 2008; 9: 75967.

    Colca JR, McDonald WG, Waldon DJ, Leone JW, Lull JM, Bannow CA,

    et al. Identification of a novel mitochondrial protein ("mitoNEET")

    cross-linked specifically by a thiazolidinedione photoprobe.

    Am J Physiol Endocrinol Metab 2004; 286: E25260.

    Cooper JM, Mann VM, Schapira AH. Analyses of mitochondrial

    respiratory chain function and mitochondrial DNA deletion

    in human skeletal muscle: effect of ageing. J Neurol Sci 1992; 113:

    918.

    Craft S, Watson GS. Insulin and neurodegenerative disease: shared and

    specific mechanisms. Lancet Neurol 2004; 3: 16978.

    Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D.Transcriptional repression of PGC-1a by mutant Huntingtin leads

    to mitochondrial dysfunction and neurodegeneration. Cell 2006; 127:

    5969.

    DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD.

    Effects of exenatide (exendin-4) on glycemic control and weight

    over 30 weeks in metformin-treated patients with type 2 diabetes.

    Diabetes Care 2005; 28: 1092100.

    Dehmer T, Heneka MT, Sastre M, Dichgans J, Schulz JB. Protection by

    pioglitazone in the MPTP model of Parkinsons disease correlates with

    I kappa B alpha induction and block of NF kappa B and iNOS activa-

    tion. J Neurochem 2004; 88: 494501.

    Devi L, Raghavendran V, Prabhu BM, Avadhani NG,

    Anandatheerthavarada HK. Mitochondrial import and accumulation

    of alpha-synuclein impair complex I in human dopaminergic neuronal

    cultures and Parkinson disease brain. J Biol Chem 2008; 283:

    9089100.

    Drucker DJ, Buse JB, Taylor K, Kendall DM, Trautmann M, Zhuang D,

    et al. Exenatide once weekly versus twice daily for the treatment of

    type 2 diabetes: a randomised, open-label, non-inferiority study.

    Lancet 2008; 372: 124050.

    Ek J, Andersen G, Urhammer SA, Gaede PH, Drivsholm T, Borch-

    Johnsen K, et al. Mutation analysis of peroxisome proliferator-

    activated receptor-gamma coactivator-1 (PGC-1) and relationships of

    identified amino acid polymorphisms to Type II diabetes mellitus.

    Diabetologia 2001; 44: 22206.

    Elner AM, Kandel EI. [Studies on the Carbohydrate metabolism in

    Parkinsonism. (Relation of Carbohydrate metabolism disorders to the

    clinical picture of the disease)]. Zh Nevropatol Psikhiatr Im S S

    Korsakova 1965; 65: 4650.

    Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and

    stress-activated signaling pathways: a unifying hypothesis of type 2diabetes. Endocr Rev 2002; 23: 599622.

    Fan R, Li X, Gu X, Chan JC, Xu G. Exendin-4 protects pancreatic beta

    cells from human islet amyloid polypeptide-induced cell damage: po-

    tential involvement of AKT and mitochondria biogenesis. Diabetes

    Obes Metab 2010; 12: 81524.

    Feany MB, Bender WW. A Drosophila model of Parkinsons disease.

    Nature 2000; 404: 3948.

    Finck BN, Kelly DP. PGC-1 coactivators: inducible regulators of

    energy metabolism in health and disease. J Clin Invest 2006; 116:

    61522.

    Finsterer J, Stollberger C, Feichtinger H. Noncompaction and endocarditis

    in suspected mitochondrial disorder. Int J Cardiol 2008; 123: e457.

    8 | Brain 2012: Page 8 of 11 I. Aviles-Olmos et al.

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    9/11

    Gerbitz KD, van den Ouweland JM, Maassen JA, Jaksch M.

    Mitochondrial diabetes mellitus: a review. Biochim Biophys Acta

    1995; 1271: 25360.

    Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al.

    In vivo imaging of microglial activation with [11C](R)-

    PK11195 PET in idiopathic Parkinsons disease. Neurobiol Dis 2006;

    21: 40412.

    Gerlach M, Riederer P, Przuntek H, Youdim MB. MPTP mechanisms of

    neurotoxicity and their implications for Parkinsons disease.

    Eur J Pharmacol 1991; 208: 27386.Ghosh S, Patel N, Rahn D, McAllister J, Sadeghi S, Horwitz G, et al. The

    thiazolidinedione pioglitazone alters mitochondrial function in human

    neuron-like cells. Mol Pharmacol 2007; 71: 1695702.

    Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM,

    Habel LA, et al. Diabetes and cancer: a consensus report. Diabetes

    Care 2010; 33: 167485.

    Greggio E, Bisaglia M, Civiero L, Bubacco L. Leucine rich repeat kinase 2

    and alpha synuclein: intersecting pathways in the pathogenesis of

    Parkinsons disease? Mol Neurodegen 2011; 6: 6.

    Guo Z, Ersoz A, Butterfield DA, Mattson MP. Beneficial effects of dietary

    restriction on cerebral cortical synaptic terminals: preservation of glu-

    cose and glutamate transport and mitochondrial function after expos-

    ure to amyloid beta-peptide, iron, and 3-nitropropionic acid. J

    Neurochem 2000; 75: 31420.

    Handschin C, Spiegelman BM. The role of exercise and PGC1alpha ininflammation and chronic disease. Nature 2008; 454: 4639.

    Hara K, Tobe K, Okada T, Kadowaki H, Akanuma Y, Ito C, et al. A

    genetic variation in the PGC-1 gene could confer insulin resistance

    and susceptibility to Type II diabetes. Diabetologia 2002; 45: 7403.

    Hara K, Yamamoto M, Anegawa T, Sakuta R, Nakamura M.

    [Mitochondrial encephalomyopathy associated with parkinsonism and

    a point mutation in the mitochondrial tRNA(Leu)(UUR)) gene]. Rinsho

    Shinkeigaku 1994; 34: 3615.

    Harkavyi A, Abuirmeileh A, Lever R, Kingsbury AE, Biggs CS,

    Whitton PS. Glucagon-like peptide 1 receptor stimulation reverses

    key deficits in distinct rodent models of Parkinsons disease. J

    Neuroinflammation 2008; 5: 19.

    Heilbronn LK, Gan SK, Turner N, Campbell LV, Chisholm DJ. Markers of

    mitochondrial biogenesis and metabolism are lower in overweight and

    obese insulin-resistant subjects. J Clin Endocrinol Metab 2007; 92:

    146773.

    Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;

    444: 8607.

    Hu G, Jousilahti P, Bidel S, Antikainen R, Tuomilehto J. Type 2 diabetes

    and the risk of Parkinsons disease. Diabetes Care 2007; 30: 8427.

    Hu G, Jousilahti P, Nissinen A, Antikainen R, Kivipelto M, Tuomilehto J.

    Body mass index and the risk of Parkinson disease. Neurology 2006;

    67: 19559.

    Hu G, Jousilahti P, Sarti C, Antikainen R, Tuomilehto J. The effect of

    diabetes and stroke at baseline and during follow-up on stroke mor-

    tality. Diabetologia 2006; 49: 230916.

    Hu MT, Taylor-Robinson SD, Chaudhuri KR, Bell JD, Labbe C,

    Cunningham VJ, et al. Cortical dysfunction in non-demented

    Parkinsons disease patients: a combined (31)P-MRS and

    (18)FDG-PET study. Brain 2000; 123: 34052.

    Hunter RL, Dragicevic N, Seifert K, Choi DY, Liu M, Kim HC, et al.Inflammation induces mitochondrial dysfunction and dopaminergic

    neurodegeneration in the nigrostriatal system. J Neurochem 2007;

    100: 137586.

    Hurlbert MS, Zhou W, Wasmeier C, Kaddis FG, Hutton JC, Freed CR.

    Mice transgenic for an expanded CAG repeat in the Huntingtons

    disease gene develop diabetes. Diabetes 1999; 48: 64951.

    Irrcher I, Aleyasin H, Seifert EL, Hewitt SJ, Chhabra S, Phillips M, et al.

    Loss of the Parkinsons disease-linked gene DJ-1 perturbs mitochon-

    drial dynamics. Hum Mol Genet 2010; 19: 373446.

    Janson J, Laedtke T, Parisi JE, OBrien P, Petersen RC, Butler PC.

    Increased Risk of Type 2 Diabetes in Alzheimer Disease. Diabetes

    2004; 53: 47481.

    Kannangara TS, Lucero MJ, Gil-Mohapel J, Drapala RJ, Simpson JM,

    Christie BR, et al. Running reduces stress and enhances cell genesis

    in aged mice. Neurobiol Aging 2011; 32: 227986.

    Kapogiannis D, Mattson MP. Disrupted energy metabolism and neuronal

    circuit dysfunction in cognitive impairment and Alzheimers disease.

    Lancet Neurol 2011; 10: 18798.

    Keeney PM, Xie J, Capaldi RA, Bennett JP Jr. Parkinsons disease

    brain mitochondrial complex I has oxidatively damaged subunits and

    is functionally impaired and misassembled. J Neurosci 2006; 26:

    525664.Kendall DM, Riddle MC, Rosenstock J, Zhuang D, Kim DD, Fineman MS,

    et al. Effects of exenatide (exendin-4) on glycemic control over 30

    weeks in patients with type 2 diabetes treated with metformin and

    a sulfonylurea. Diabetes Care 2005; 28: 108391.

    Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue

    tumor necrosis factor and interleukin-6 expression in human obesity

    and insulin resistance. Am J Physiol Endocrinol Metab 2001; 280:

    E74551.

    Kim Chung le T, Hosaka T, Yoshida M, Harada N, Sakaue H, Sakai T,

    et al. Exendin-4, a GLP-1 receptor agonist, directly induces adiponectin

    expression through protein kinase A pathway and prevents inflamma-

    tory adipokine expression. Biochem Biophys Res Commun 2009; 390:

    6138.

    Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, et al.

    Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci

    USA 2005; 102: 521520.

    Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by

    inhibition of microglial activation and matrix metalloproteinase-3 ex-

    pression in an animal model of Parkinsons disease. J Endocrinol 2009;

    202: 4319.

    Kraytsberg Y, Kudryavtseva E, McKee AC, Geula C, Kowall NW,

    Khrapko K. Mitochondrial DNA deletions are abundant and cause

    functional impairment in aged human substantia nigra neurons. Nat

    Genet 2006; 38: 51820.

    LaManna JC, Harik SI. Regional comparisons of brain glucose influx.

    Brain Res 1985; 326: 299305.

    Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in

    humans due to a product of meperidine-analog synthesis. Science

    1983; 219: 97980.

    Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, OBrien PC, et al.

    Risk of dementia among persons with diabetes mellitus: a

    population-based cohort study. Am J Epidemiol 1997; 145: 3018.

    Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, et al.

    GLP-1 receptor stimulation preserves primary cortical and dopamin-

    ergic neurons in cellular and rodent models of stroke and

    Parkinsonism. Proc Natl Acad Sci USA 2009; 106: 128590.

    Li Y, Tweedie D, Mattson MP, Holloway HW, Greig NH. Enhancing the

    GLP-1 receptor signaling pathway leads to proliferation and neuropro-

    tection in human neuroblastoma cells. J Neurochem 2010; 113:

    162131.

    Lin J, Handschin C, Spiegelman BM. Metabolic control through the

    PGC-1 family of transcription coactivators. Cell Metab 2005; 1:

    36170.

    Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, et al. Transcriptional

    co-activator PGC-1 alpha drives the formation of slow-twitch musclefibres. Nature 2002; 418: 797801.

    Lipman IJ, Boykin ME, Flora RE. Glucose intolerance in Parkinsons dis-

    ease. J Chronic Dis 1974; 27: 5739.

    Luchsinger JA, Reitz C, Patel B, Tang MX, Manly JJ, Mayeux R. Relation

    of diabetes to mild cognitive impairment. Arch Neurol 2007; 64:

    5705.

    Luoma PT, Eerola J, Ahola S, Hakonen AH, Hellstrom O, Kivisto KT, et al.

    Mitochondrial DNA polymerase gamma variants in idiopathic sporadic

    Parkinson disease. Neurology 2007; 69: 11529.

    Mann VM, Cooper JM, Daniel SE, Srai K, Jenner P, Marsden CD, et al.

    Complex I, iron, and ferritin in Parkinsons disease substantia nigra.

    Ann Neurol 1994; 36: 87681.

    Parkinsons disease and insulin resistance Brain 2012: Page 9 of 11 | 9

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    10/11

    Matsuda W, Furuta T, Nakamura KC, Hioki H, Fujiyama F, Arai R, et al.

    Single nigrostriatal dopaminergic neurons form widely spread and

    highly dense axonal arborizations in the neostriatum. J Neurosci

    2009; 29: 44453.

    Mattson MP, Gleichmann M, Cheng A. Mitochondria in neuroplasticity

    and neurological disorders. Neuron 2008; 60: 74866.

    Mazzulli JR, Xu YH, Sun Y, Kinght AL, McLean PJ, Caldwell GA, et al.

    Gaucher disease glucocerebrosidase and -synuclein form a

    bidirectional pathogenic loop in synucleinopathies. Cell 2011; 146:

    911.McFarland MA, Ellis CE, Markey SP, Nussbaum RL. Proteomics analysis

    identifies phosphorylation-dependent alpha-synuclein protein inter-

    actions. Mol Cell Proteomics 2008; 7: 212337.

    McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are

    positive for HLA-DR in the substantia nigra of Parkinsons and

    Alzheimers disease brains. Neurology 1988; 38: 128591.

    Montefusco O, Assini MC, Missale C. Insulin-mediated effects of glucose

    on dopamine metabolism. Acta Diabetol Lat 1983; 20: 717.

    Moroo I, Yamada T, Makino H, Tooyama I, McGeer PL, McGeer EG,

    et al. Loss of insulin receptor immunoreactivity from the substantia

    nigra pars compacta neurons in Parkinsons disease. Acta

    Neuropathol 1994; 87: 3438.

    Moss J, Bolam JP. The relationship between dopaminergic axons and

    glutamatergic synapses in the striatum. New York: Oxford University

    Press; 2010.Mounier C, Posner BI. Transcriptional regulation by insulin: from the

    receptor to the gene. Can J Physiol Pharmacol 2006; 84: 71324.

    Muftuoglu M, Elibol B, Dalmizrak O, Ercan A, Kulaksiz G, Ogus H, et al.

    Mitochondrial complex I and IV activities in leukocytes from patients

    with parkin mutations. Mov Disord 2004; 19: 5448.

    Murzi E, Contreras Q, Teneud L, Valecillos B, Parada MA, De

    Parada MP, et al. Diabetes decreases limbic extracellular dopamine

    in rats. Neurosci Lett 1996; 202: 1414.

    Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin UM, Saad M,

    et al. Imputation of sequence variants for identification of genetic risks

    for Parkinsons disease: a meta-analysis of genome-wide association

    studies. Lancet 2011; 377: 6419.

    Orr AL, Li S, Wang CE, Li H, Wang J, Rong J, et al. N-terminal mutant

    huntingtin associates with mitochondria and impairs mitochondrial

    trafficking. J Neurosci 2008; 28: 278392.

    Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM.

    Diabetes mellitus and the risk of dementia: The Rotterdam Study.

    Neurology 1999; 53: 193742.

    Paddock ML, Wiley SE, Axelrod HL, Cohen AE, Roy M, Abresch EC,

    et al. MitoNEET is a uniquely folded 2Fe 2S outer mitochondrial mem-

    brane protein stabilized by pioglitazone. Proc Natl Acad Sci USA 2007;

    104: 143427.

    Palacios N, Gao X, McCullough ML, Jacobs EJ, Patel AV, Mayo T, et al.

    Obesity, diabetes, and risk of Parkinsons disease. Mov Disord 2011;

    26: 22539.

    Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, Wacker M, et al.

    Mitochondrial dysfunction and oxidative damage in Parkin-deficient

    mice. J Biol Chem 2004; 279: 1861422.

    Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, et al.

    Coordinated reduction of genes of oxidative metabolism in humans

    with insulin resistance and diabetes: potential role of PGC1 andNRF1. Proc Natl Acad Sci USA 2003; 100: 846671.

    Peila R, Rodriguez BL, Launer LJ. Type 2 diabetes, APOE gene, and the

    risk for dementia and related pathologies: The Honolulu-Asia Aging

    Study. Diabetes 2002; 51: 125662.

    Perier C, Tieu K, Guegan C, Caspersen C, Jackson-Lewis V, Carelli V,

    et al. Complex I deficiency primes Bax-dependent neuronal apoptosis

    through mitochondrial oxidative damage. Proc Natl Acad Sci USA

    2005; 102: 1912631.

    Perry TA, Greig NH. A new Alzheimers disease interventive strategy:

    GLP-1. Curr Drug Targets 2004; 5: 56571.

    Perry T, Lahiri DK, Chen D, Zhou J, Shaw KT, Egan JM, et al. A novel

    neurotrophic property of glucagon-like peptide 1: a promoter of nerve

    growth factor-mediated differentiation in PC12 cells. J Pharmacol Exp

    Ther 2002; 300: 95866.

    Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mito-

    chondrial activity in the insulin-resistant offspring of patients with type

    2 diabetes. N Engl J Med 2004; 350: 66471.

    Pistell PJ, Morrison CD, Gupta S, Knight AG, Keller JN, Ingram DK, et al.

    Cognitive impairment following high fat diet consumption is associated

    with brain inflammation. J Neuroimmunol 2010; 219: 2532.

    Podolsky S, Leopold NA, Sax DS. Increased frequency of diabetes melli-

    tus in patients with Huntingtons chorea. Lancet 1972; 1: 13568.Poon HF, Frasier M, Shreve N, Calabrese V, Wolozin B, Butterfield DA.

    Mitochondrial associated metabolic proteins are selectively oxidized in

    A30P alpha-synuclein transgenic micea model of familial Parkinsons

    disease. Neurobiol Dis 2005; 18: 4928.

    Pressley JC, Louis ED, Tang MX, Cote L, Cohen PD, Glied S, et al. The

    impact of comorbid disease and injuries on resource use and expend-

    itures in Parkinsonism. Neurology 2003; 60: 8793.

    Pyle A, Foltynie T, Tiangyou W, Lambert C, Keers SM, Allcock LM, et al.

    Mitochondrial DNA haplogroup cluster UKJT reduces the risk of PD.

    Ann Neurol 2005; 57: 5647.

    Qin W, Haroutunian V, Katsel P, Cardozo CP, Ho L, Buxbaum JD, et al.

    PGC-1 Expression decreases in the Alzheimer disease brain as a func-

    tion of dementia. Arch Neurol 2009; 66: 35261.

    Quinn LP, Crook B, Hows ME, Vidgeon-Hart M, Chapman H, Upton N,

    et al. The PPARgamma agonist pioglitazone is effective in the MPTPmouse model of Parkinsons disease through inhibition of monoamine

    oxidase B. Br J Pharmacol 2008; 154: 22633.

    Rakovic A, Grunewald A, Seibler P, Ramirez A, Kock N, Orolicki S, et al.

    Effect of endogenous mutant and wild-type PINK1 on Parkin in fibro-

    blasts from Parkinson disease patients. Hum Mol Genet 2010; 19:

    312437.

    Ramonet D, Paulo J, Daher L, Lin BM, Stafa K, Kim J, et al.

    Dopaminergic neuronal loss, reduced neurite complexity and autopha-

    gic abnormalities in transgenic mice expressing G2019S mutant

    LRRK2. PLoS One 2011; 6: e18568.

    Ren H, Fu K, Wang D, Mu C, Wang G. Oxidised DJ-1 interacts with the

    mitochondrial protein BCL-XL. J Biol Chem 2011; 286: 3530817.

    Ristow M. Neurodegenerative disorders associated with diabetes mellitus.

    J Mol Med 2004; 82: 51029.

    Saller CF, Chiodo LA. Glucose suppresses basal firing and

    haloperidol-induced increases in the firing rate of central dopaminergic

    neurons. Science 1980; 210: 126971.

    Sandyk R. The relationship between diabetes mellitus and Parkinsons

    disease. Int J Neurosci 1993; 69: 12530.

    Schapira AH. Mitochondria in the aetiology and pathogenesis of

    Parkinsons disease. Lancet Neurol 2008; 7: 97109.

    Schapira AH. Complex I: inhibitors, inhibition and neurodegeneration.

    Exp Neurol 2010; 224: 3315.

    Schapira AH, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD.

    Mitochondrial complex I deficiency in Parkinsons disease. Lancet

    1989; 1: 1269.

    Schwab RS. Progression and prognosis in Parkinsons disease. J Nerv

    Ment Dis 1960; 130: 55666.

    Shin JH, Ko HS, Kang H, Lee Y, Lee YI, Pletinkova O, et al. PARIS

    (ZNF746) repression of PGC-1alpha contributes to neurodegeneration

    in Parkinsons disease. Cell 2011; 144: 689702.Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER,

    et al. Multicenter analysis of glucocerebrosidase mutations in

    Parkinsons disease. N Engl J Med 2009; 361: 165161.

    Simon DK, Pulst SM, Sutton JP, Browne SE, Beal MF, Johns DR.

    Familial multisystem degeneration with parkinsonism associated with

    the 11778 mitochondrial DNA mutation. Neurology 1999; 53:

    178793.

    Sirtori CR, Bolme P, Azarnoff DL. Metabolic responses to acute and

    chronic L-dopa administration in patients with parkinsonism. N Engl

    J Med 1972; 287: 72933.

    Staiger H, Stefan N, Machicao F, Fritsche A, Haring HU. PPARGC1A

    mRNA levels of in vitro differentiated human skeletal muscle cells

    10 | Brain 2012: Page 10 of 11 I. Aviles-Olmos et al.

  • 7/30/2019 Brain 2012 Aviles Olmos Brain Aws009 (1)

    11/11

    are negatively associated with the plasma oleate concentrations of the

    donors. Diabetologia 2006; 49: 2124.

    St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager S, et al.

    Suppression of reactive oxygen species and neurodegeneration by

    the PGC-1 transcriptional coactivators. Cell 2006; 127: 397408.

    Stark R, Roden M. ESCI Award 2006. Mitochondrial function and endo-

    crine diseases. Eur J Clin Invest 2007; 37: 23648.

    Stichel CC, Augustin M, Kuhn K, Zhu XR, Engels P, Ullmer C, et al.

    Parkin expression in the adult mouse brain. Eur J Neurosci 2000; 12:

    418194.Stranahan AM, Arumugam TV, Cutler RG, Lee K, Egan JM, Mattson MP.

    Diabetes impairs hippocampal function through glucocorticoid-

    mediated effects on new and mature neurons. Nat Neurosci 2008;

    11: 30917.

    Stranahan AM, Arumugam TV, Lee K, Mattson MP. Mineralocorticoid

    receptor activation restores medial perforant path LTP in diabetic rats.

    Synapse 2010; 64: 52832.

    Summers SA, Nelson DH. A role for Sphingolipids in producing the

    common features of Type 2 Diabetes, Metabolic Syndrome X, and

    Cushings syndrome. Diabetes 2005; 54: 591602.

    Takahashi M, Yamada T, Tooyama I, Moroo I, Kimura H, Yamamoto T,

    et al. Insulin receptor mRNA in the substantia nigra in Parkinsons

    disease. Neurosci Lett 1996; 204: 2014.

    Takeda S, Sato N, Uchio-Yamada K, Sawada K, Kunieda T, Takeuchi D,

    et al. Diabetes-accelerated memory dysfunction via cerebrovascular

    inflammation and Abeta deposition in an Alzheimer mouse model

    with diabetes. Proc Natl Acad Sci USA 2010; 107: 703641.

    Tansey MG, Frank-Cannon TC, McCoy MK, Lee JK, Martinez TN,

    McAlpine FE, et al. Neuroinflammation in Parkinsons disease: is

    there sufficient evidence for mechanism-based interventional therapy?

    Front Biosci 2008; 13: 70917.

    Toro P, Schonknecht P, Schroder J. Type II diabetes in mild cognitive

    impairment and Alzheimers disease: results from a prospective

    population-based study in Germany. J Alzheimers Dis 2009; 16:

    68791.

    Tuomilehto J, Rastenyte D, Jousilahti P, Sarti C, Vartiainen E. Diabetes

    mellitus as a risk factor for death from stroke. Prospective study of the

    middle-aged Finnish population. Stroke 1996; 27: 2105.

    Unger JW, Livingston JN, Moss AM. Insulin receptors in the central ner-

    vous system: localization, signalling mechanisms and functional as-

    pects. Prog Neurobiol 1991; 36: 34362.

    Van Woert MH, Mueller PS. Glucose, insulin, and free fatty acid metab-

    olism in Parkinsons disease treated with levodopa. Clin Pharmacol

    Ther 1971; 12: 3607.

    Vives-Bauza C, Przedborski S. Mitophagy: the latest problem for

    Parkinsons disease. Trends Mol Med 2011; 17: 15865.

    Wahner AD, Bronstein JM, Bordelon YM, Ritz B. Nonsteroidal

    anti-inflammatory drugs may protect against Parkinson disease.

    Neurology 2007; 69: 183642.

    Wang X, Su B, Siedlak SL, Moreira PI, Fujioka H, Wang Y, et al.

    Amyloid-b overproduction causes abnormal mitochondrial dynamicsvia differential modulation of mitochondrial fission/fusion proteins.

    Proc Natl Acad Sci USA 2008; 105: 1931823.

    Westbroek W, Gustafson AM, Sidransky E. Exploring the link between

    glucocerebrosidase mutations and parkinsonism. Trends Mol Med

    2011; 17: 48593.

    Whitton PS. Inflammation as a causative factor in the aetiology of

    Parkinsons disease. Br J Pharmacol 2007; 150: 96376.

    Wiley SE, Paddock ML, Abresch EC, Gross L, van der Geer P,

    Nechushtai R, et al. The outer mitochondrial membrane protein

    mitoNEET contains a novel redox-active 2Fe-2S cluster. J Biol Chem

    2007; 282: 237459.

    Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative dis-

    eases. Nat Neurosci 2010; 13: 80511.

    Wu W, Brickman AM, Luchsinger J, Ferrazzano P, Pichiule P, Yoshita M,

    et al. The brain in the age of old: the hippocampal formation is tar-geted differentially by diseases of late life. Ann Neurol 2008; 64:

    698706.

    Yokota T, Sugawara K, Ito K, Takahashi R, Ariga H, Mizusawa H. Down

    regulation of DJ-1 enhances cell death by oxidative stress, ER stress,

    and proteasome inhibition. Biochem Biophys Res Commun 2003; 312:

    13428.

    Zhang L, Shimoji M, Thomas B, Moore DJ, Yu SW, Marupudi NI, et al.

    Mitochondrial localization of the Parkinsons disease related protein

    DJ-1: implications for pathogenesis. Hum Mol Genet 2005; 14:

    206373.

    Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, et al.

    PGC-1alpha, a potential therapeutic target for early intervention in

    Parkinsons disease. Sci Transl Med 2010; 2: 5273.

    Parkinsons disease and insulin resistance Brain 2012: Page 11 of 11 | 11