page 1 of 45 diabetes · epc and lks cells after ischemia, epc levels peaked at day 3 after...

45
1 Diabetes causes bone marrow autonomic neuropathy and impairs stem cell mobilization via dysregulated p66Shc and Sirt1 Running title “Autonomic neuropathy and stem cell mobilopathy” Authors Mattia Albiero 1 , Nicol Poncina 1,2 , Marc Tjwa 3 , Stefano Ciciliot 1,2 , Lisa Menegazzo 1,2 ,Giulio Ceolotto 2 , Saula Vigili de Kreutzenberg 2 , Rute Moura 3 , Marco Giorgio 4 , Piergiuseppe Pelicci 4 , Angelo Avogaro 1,2 , and Gian Paolo Fadini 1,2 Affiliations 1 Venetian Institute of Molecular Medicine, 35100 Padova, Italy. 2 Department of Medicine, University of Padova, 35100 Padova, Italy 3 Laboratory Vascular Hematology / Angiogenesis, Institute for Transfusion Medicine, Goethe University Frankfurt, 60528 Frankfurt, Germany. 4 European Institute of Oncology, IFOM-IEO Campus, 20139 Milan, Italy. Corresponding author Dr. Gian Paolo Fadini Department of Medicine, University of Padova. Via Giustiniani, 2. 35100 Padova. Phone +39 049 8214318 Fax +39 049 8212184 Email: [email protected]; [email protected] Word count 4276 1 Tables 6 Figures The manuscript has Online Supplemental Material Page 1 of 45 Diabetes Diabetes Publish Ahead of Print, published online November 22, 2013

Upload: others

Post on 04-Jun-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

1

Diabetes causes bone marrow autonomic neuropathy

and impairs stem cell mobilization via dysregulated p66Shc and Sirt1

Running title “Autonomic neuropathy and stem cell mobilopathy”

Authors Mattia Albiero

1, Nicol Poncina

1,2, Marc Tjwa

3, Stefano Ciciliot

1,2, Lisa Menegazzo

1,2,Giulio

Ceolotto2, Saula Vigili de Kreutzenberg

2, Rute Moura

3, Marco Giorgio

4, Piergiuseppe

Pelicci4, Angelo Avogaro

1,2, and Gian Paolo Fadini

1,2

Affiliations 1 Venetian Institute of Molecular Medicine, 35100 Padova, Italy.

2 Department of Medicine, University of Padova, 35100 Padova, Italy

3 Laboratory Vascular Hematology / Angiogenesis, Institute for Transfusion Medicine,

Goethe University Frankfurt, 60528 Frankfurt, Germany. 4 European Institute of Oncology, IFOM-IEO Campus, 20139 Milan, Italy.

Corresponding author

Dr. Gian Paolo Fadini

Department of Medicine, University of Padova.

Via Giustiniani, 2. 35100 Padova.

Phone +39 049 8214318

Fax +39 049 8212184

Email: [email protected];

[email protected]

Word count 4276 1 Tables

6 Figures

The manuscript has Online Supplemental Material

Page 1 of 45 Diabetes

Diabetes Publish Ahead of Print, published online November 22, 2013

Page 2: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

2

ABSTRACT

Diabetes compromises the bone marrow (BM) microenvironment and reduces circulating

CD34+ cells. Diabetic autonomic neuropathy (DAN) may impact the BM, because the

sympathetic nervous system (SNS) is prominently involved in BM stem cell trafficking. We

hypothesize that neuropathy of the BM affects stem cell mobilization and vascular recovery

after ischemia in diabetes. We report that, in patients, cardiovascular DAN was associated

with fewer circulating CD34+ cells. Experimental diabetes (STZ and Ob/Ob) or chemical

sympathectomy in mice resulted in BM autonomic neuropathy, impaired Lin-cKit

+Sca1

+

(LKS) cell and endothelial progenitor cells (EPC, CD34+Flk1

+) mobilization and vascular

recovery after ischemia. DAN increased expression of p66Shc and reduced expression of

Sirt1 in mice and humans. p66Shc KO in diabetic mice prevented DAN in the BM, and

rescued defective LKS cell and EPC mobilization. Hematopoietic Sirt1 KO mimicked the

diabetic mobilization defect, while hematopoietic Sirt1 overexpression in diabetes rescued

defective mobilization and vascular repair. Through p66Shc and Sirt1, diabetes and

sympathectomy elevated the expression of various adhesion molecules, including CD62L.

CD62L KO partially rescued the defective stem/progenitor cell mobilization. In conclusion,

autonomic neuropathy in the BM impairs stem cell mobilization in diabetes with

dysregulation of the lifespan regulators p66Shc and Sirt1.

Page 2 of 45Diabetes

Page 3: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

3

Diabetes reduces the availability of bone marrow (BM)-derived circulating angiocompetent

CD34+ cells, especially in the presence of chronic vascular complications (1; 2). This is

believed to represent a risk factor for adverse outcomes, as low CD34+ cell count is associated

with incident cardiovascular events (3). Recently, the BM has emerged as a previously

neglected target of hyperglycaemic damage in experimental and human diabetes, with

features of microangiopathy and niche dysfunction (4-7). As a result of BM remodelling,

mobilization of stem and progenitor cells is impaired in diabetic animals and humans (7-10).

Since mobilization of BM-derived stem/progenitor cells contributes to angiogenesis and

endothelial repair (11; 12), such stem cell “mobilopathy” (13) is expected to promote

cardiovascular disease in diabetes.

The sympathetic nervous system (SNS) is prominently involved in BM niche function (14)

and stem cell trafficking is regulated by catecholaminergic neurotransmitters (15; 16).

Recently, it has been also demonstrated that BM denervation strongly contributes to

chemotherapy-induced myelotoxicity (17). Diabetic patients often suffer from autonomic

neuropathy, characterized by pauperization of SNS fibers in multiple organs (18-20). Thus,

we herein hypothesize that diabetic autonomic neuropathy (DAN) can affect BM function and

stem cell mobilization. Starting from the observation that patients with DAN have a marked

pauperization of peripheral blood CD34+ cells, we used several experimental models and

conditions to demonstrate that autonomic neuropathy in the BM is causally linked to impaired

stem cell mobilization in diabetes, associated with defective reperfusion after ischemia. Our

data show that denervation in the diabetic BM is mediated by p66Shc and that impaired

mobilization relies on Sirt1 dysregulation, both of which are therefore potential targets to

restore BM-cell mediated vascular repair.

Page 3 of 45 Diabetes

Page 4: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

4

RESEARCH DESIGN AND METHODS

Patients. The protocol was approved by the Ethics Committee of the University Hospital of

Padova and experiments were conducted in accordance to the Declaration of Helsinki as

revised in 2000. Patients were recruited at the diabetes outpatient clinic of the University

Hospital of Padova. All consecutive type 1 or type 2 diabetic patients aged 20-90 and

undergoing autonomic neuropathy tests were eligible, provided they were clinically stable and

free from exclusion criteria (recent acute disease or infection, trauma or surgery; uncontrolled

hypertension or hyperglycemia, prevalent hypoglycaemia; immunological disorders, organ

transplantation or immunosuppression; pregnancy and lactation). The following data were

recorded: age sex, type of diabetes and disease duration, HbA1c, BMI, waist circumference,

smoking habit, systolic and diastolic blood pressure, history of hypertension of use of anti-

hypertensive medications, resting heart rate, lipid profile, albumin/creatinine excretion ratio,

diabetic complications and medications. Diabetic nephropathy was defined as an urinary

albumin/creatinine ratio >30 mg/g or an estimated glomerular filtration rate (eGFR) <60

mL/min/1.73 m2. Coronary artery disease was defined as a past history of myocardial

infarction or angina, confirmed by coronary artery angiography or a myocardial stress test.

Peripheral arterial disease was defined as a history of claudication or rest pain with evidence

of stenosis of leg arteries at invasive or non invasive examinations. Cerebrovascular disease

was defined as a history of stroke or evidence of >30% carotid stenosis at ultrasound

examination. Diagnosis of DAN was performed according to standard interpretation of R-R

interval variations during lying-to-standing, deep breathing, Valsalva manoeuvre, as well as

according to the degree of orthostatic hypotension. Age-specific thresholds were used to

define pathological versus normal DAN tests.

Page 4 of 45Diabetes

Page 5: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

5

Animals. All procedures were approved by the local ethic committee and from the Italian

Ministry of Health. Experiments were conducted according to the “Principles of laboratory

animal care” (NIH publication no. 85–23,4 revised 1985). Vav1-Sirt1-/- (generated by crossing

Vav1cre/+

mice with Sirt1stop-flox

mice), Vav1-Sirt1TG (generated by crossing

Vav1cre/+

Sirt1ex4.∆flox/+

with Sirt1ex4.flox/flox

) and Vav1-YPF were bred at the external mouse

facilities of the University of Frankfurt, MFD Diagnostics GmbH. Sell-/-

mice were bred at the

mouse facilities of the Institut für Labortierkunde, University of Zurich, Zurich, Switzerland.

Sell-/-

mice were generated by Richard Hynes, a Howard Hughes Medical Institute ("HHMI")

Investigator at Massachusetts Institute of Technology. p66Shc-/-

and Ob/Ob p66Shc-/-

animals

were bred at the animal facility of the University of Padova. Ob/Ob mice were from Jackson

Laboratory (Maine, USA). Age-matched wild-type animals were obtained from the in-house

colony of the animal facility of the Venetian Institute of Molecular Medicine. All animals

were on a C57BL/6 background. Diabetes was induced with a single i.p. injection of 150

mg/kg STZ (Sigma Aldrich, St.Louis, MO, USA) in citrate buffer 50 mM, ph 4.5. Blood

glucose was measured with Glucocard G-meter (Menarini, Florence, Italy); animals with

blood glucose ≥300 mg/dl in at least two measurements within the first week were classified

as diabetic and housed for 4 weeks with feeding and drinking ad libitum before performing

experiments. In a separate set, animals were sacrificed at week 1, 2, and 3 after STZ treatment

for a time-course analysis (n=4-5/group). For SNS disruption, animals received 3

intraperitoneal injections 100 mg/kg 6-hydroxy-dopamine (6-OHDA, Tocris Bioscience,

Bristol, UK), which destroys sympathetic neurons, through oxidative stress and other

mechanisms. Animals were housed for 4 weeks before a second round of injections with 2

doses of 6-OHDA (100 mg/kg on day 0, 250 mg/kg on day 2), and experiments were started

on day 5.

Page 5 of 45 Diabetes

Page 6: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

6

Hind limb ischemia. Animals were sedated with 10 mg/kg zolazepam/thylamine (Zoletil®,

Laboratories Virbac, Nice, France) and 7 mg/kg xylazine (Xilor®, Laboratories Carlier,

Spain). The femoral artery and the vein were surgically dissected from the femoral nerve, then

cauterized with low temperature cautery and excised between inguinal ligament and hackle.

Fifteen days after surgery, we measured hind limb microvascular perfusion with Perimed

Periscan-Pim II Laser Doppler System (Perimed AB, Sweden). Each measure was repeated 5

times.

Mobilization assays and flow cytometry. Progenitor cell levels were quantified in peripheral

blood after peripheral ischemia or after 4 days of s.c. injection of 200 µg/kg/die of hrG-CSF

(Filgrastim, Roche, Basel, Switzerland). Based on a preliminary analysis of the kinetics of

EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS

peaked at day 14. Therefore, these time points were used for all subsequent analyses.

AMD3100 (Tocris) was given i.p. at 6 mg/kg and mobilization assessed after 4 hours.

Desipramine treatment (10 mg/kg i.p., Tocris) was started 4 days before G-CSF and continued

for the 4 days of G-CSF treatment. Peripheral blood was collected at baseline and after

mobilization. 150 µl of peripheral blood were stained with rat anti-mouse APC-lineage

cocktail (BD, NJ, USA), PE rat anti-mouse Sca-1 (Ly6A/E, BD) and FITC rat anti-mouse

cKit (BD) to quantify LKS cells or with Alexa647 rat anti-mouse CD34 (BD) and Alexa488

anti-mouse Flk-1 (Biolegend, San Diego, CA, USA) to quantify endothelial-committed

progenitors. CD62L (L-selectin) expression on bone marrow LKS cells was performed by

staining 106 non-fractioned bone marrow cells with LKS antibodies and PerCp Cy5.5 anti-

Mouse CD62L (BD). A total of 250.000 events were acquired for each analysis and the level

of progenitor cells was expressed as number of positive events per 1.000.000 total events.

Data were acquired using a FACS Calibur instrument (BD Biosciences) and analysed with

Page 6 of 45Diabetes

Page 7: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

7

FlowJo X (TreeStar Inc., USA). For quantification of circulating stem cells in humans, we

stained 150 µL of freshly collected peripheral blood with a PE-conjugated anti-human CD34

mAb (BD) and gated CD34+ cells in the mononuclear cell fraction. CD34

+ cell count was

expressed either as number of events/106 total events or cells/mL by multiplying fractional

CD34+ cell count by WBC obtained from the automated hemocytometer.

Histology. Adductor femoris longus muscles were frozen in isopentane cooled in liquid

nitrogen immediately after dissection. Muscles were then stored at -80°C for further analysis.

Muscle sections were incubated with a DyLight594-conjugated Isolectin B4 (Bandeiraea

simplicifolia, Vector Laboratories, 1:100), and co-stained with Oregon green-conjugated

wheat germ agglutinin (WGA, 1:400, Invitrogen), to allow identification of capillaries and

muscle fibers. Nuclei were counterstained with Hoechst 33258 (Sigma). Femurs were fixed

for 24h in 4% PFA at 4°C, washed with PBS, and then decalcified by incubation with 5%

formic acid for 2 days at 4°C. Demineralized femurs were then covered with OCT embedding

medium and frozen in liquid nitrogen. Ten µm thick longitudinal femur or transversal muscle

sections were obtained with a Leica CM1950 cryostat. Femur sections were fixed in 4% PFA

and incubated with tyrosine hydroxylase antibody overnight at 4°C (rabbit polyclonal, 1:100,

AB152 Millipore). A mock incubation, without primary antibody, was performed in parallel.

Images were acquired with a Leica DM5000B microscope, equipped with a Leica DFC300

FX charge-coupled device camera, and processed with ImageJ software (NIH, USA). 10

random 20x magnification pictures were obtained from each muscle sample.

In vitro experiments. Human PBMC were obtained from healthy donors and separated by

gradient centrifugation with Histopaque-1077 (Sigma). 107 cells/well were plated in 6-well

culture plate (BD) with RPMI-1640 supplemented with 10% FBS and 1%

Page 7 of 45 Diabetes

Page 8: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

8

penicillin/streptomycin/glutamine. Isopreteronol (Sigma) was added for 48 hours. Then cells

were collected, and lysed for gene expression analysis. To test chemical toxicity, BM cells

from control mice were incubated for 1h with STZ and 6-OHDA, followed by 24h of

recovery at 37°C 5% CO2 with RPMI-1640 supplemented with 10% FBS and 1%

penicillin/streptomycin/glutamine, at concentrations that are supposed to be achieved in vivo

in mice. Live, early and late apoptotic and necrotic cells (total BM cells or gated CD34+ cells)

were quantified by flow cytometry staining with propidium iodine and Annexin-V (BD).

Hematopoietic colonies were grown from unfractioned BM cells and quantified using the

Methocult system (Stem Cells inc. Vancouver, Canada).

Molecular biology. At sacrifice, bones were harvested, cleaned from debris and flushed with

ice-cold PBS; cells were collected through a 40 µm sterile filter and total RNA was extracted

using RNeasy kit (Qiagen) and RNA quantified on a Nanodrop Spectrometer (Termo Fisher

Scientific Inc). 400 ng of RNA were reverse transcribed to cDNA using the First-Strand

cDNA Synthesis Kit (Invitrogen). Duplicates of sample cDNA were then amplified on the

7900HT Fast Real-Time PCR System (Applied Biosystems) using the Fast SYBR Green RT-

PCR kit (Applied Biosystems) in 96-wells plates (Applied Biosystems). Expression data of

selected genes were normalized against housekeeping gene and were analyzed using the 2(-

∆∆CT ) method described by Livak and Schmittgen. Primer sequences are reported in online

table 3.

Statistical analysis. Data are expressed as mean ± standard error or as percentage.

Comparison between 2 or more groups was performed using Student’s t or ANOVA (for

normal variables) and with Mann-Whitney’s U and Kruskall-Wallis tests (for non-normal

variables). For human studies, a multivariable analysis was run to detect the independent

Page 8 of 45Diabetes

Page 9: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

9

effect of DAN: CD34+ cell count was the dependent variable and clinical characteristics that

were different at univariate analysis in Table 1 were explanatory independent variables.

Statistical significance was accepted at p<0.05.

Page 9 of 45 Diabetes

Page 10: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

10

RESULTS

Autonomic neuropathy impairs bone marrow stem cell mobilization in diabetes.

First, we found that the co-existence of multiple pathologic cardiovascular autonomic tests

used to diagnose DAN was associated with a progressive reduction of CD34+ cells in

peripheral blood (PB) of diabetic patients (Fig. 1A,B). Independently of confounders, patients

with DAN had a 40% reduction in PB CD34+ cells compared to DAN-free patients (Fig. 1C-

D, Table 1 and multivariate analysis in Online Table 1). A breakdown analysis by DAN test

showed trend reductions of PB CD34+ cells in the presence of alteration of each of the

autonomic function test (Fig. 1E,F). These data suggest that autonomic neuropathy in diabetic

patients reduces circulating CD34+ cells, which have been previously shown to exert pro-

angiogenic activity in vivo (21), their levels are informative of the bone marrow status (8),

and predict future cardiovascular events (22).

Next, we report that experimental T1D induced by streptozotocin (STZ) caused a marked

reduction in tyrosine-hydroxilase+ (Tyr-OH

+) SNS fibers in the BM. This was equal to about

80% of Tyr-OH staining reduction obtained by chemical sympathectomy with 6-OHDA. STZ

is considered to exert little or no myelosuppression because it enters the cells via glucose

transporters (23). However, to rule out that BM sympathectomy was attributable to STZ

toxicity and not to diabetes, we analysed BM Tyr-OH staining in Ob/Ob mice, a model of

T2D, and found a similar pauperization of SNS fibers (Fig. 2).

To test the consequence of sympathectomy on BM function in diabetes, we analysed Lin-

cKit+Sca1

+ (LKS) and CD34

+Flk1

+ cells (EPC) in T1D mice and 6-OHDA treated mice

before and after stimulation with G-CSF or hind limb ischemia. Consistent with previous

findings (8), diabetic animals were unable to mobilize LKS cells and EPC after G-CSF or

ischemia. Mice treated with 6-OHDA also showed impaired mobilization of LKS cells and

Page 10 of 45Diabetes

Page 11: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

11

EPC in response to both stimuli (Fig. 3A,B). While the SNS-dependency of the mobilizing

effect of G-CSF is well established (14), the role of BM SNS in ischemia-induced

mobilization was hitherto unknown. The BM content of EPCs, but not of LKS cells, was

reduced in diabetic animals, while steady-state circulating levels of EPC and LKS cells were

unaffected by diabetes or sympathectomy (online Fig. 1). In vitro, STZ and 6-OHDA induced

minor changes in survival of total BM cells, CD34+ BM cells and in the generation of

hematopoietic colonies (Online Fig. 2). These data rule out that chemical toxicity or absolute

pauperization of intra-marrow stem cells account for the diabetes- and neuropathy-associated

mobilopathy.

Since sympathectomy caused similar mobilization impairment as diabetes, we reasoned that

the diabetic stem cell mobilopathy could be attributed to autonomic denervation. In support of

this, we show that diabetic mice were able to mobilize LKS cells in response to

AMD3100/Plerixafor (Fig. 3C), which is a SNS-independent mobilizer (24). In addition, the

norepinephrine reuptake inhibitor desipramine, which potentiates residual SNS outflow in

autonomic ganglia (25), partially restored G-CSF-induced LKS cell mobilization in diabetic

mice (Fig. 3D).

As a functional readout of impaired mobilization after ischemia, we found that, although the

microvasculature was mildly affected, hind paw perfusion was significantly impaired in

diabetic and sympathectomised mice (Fig. 3E). This is in compliance with the known

reperfusion defect of diabetes (26; 27) and the recent finding that catecholamines regulate

post-ischemic tissue remodelling (28). Collectively, these data indicate that diabetes and

autonomic neuropathy hamper BM stem cell mobilization and post-ischemic responses.

p66Shc mediates diabetic bone marrow neuropathy.

Page 11 of 45 Diabetes

Page 12: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

12

Previous studies in mice show that p66Shc mediates hyperglycemic damage and diabetic

complications (29). Therefore, we tested whether p66Shc is involved in BM denervation

induced by diabetes. First, we report that gene expression of p66Shc was increased in PBMC

of diabetic patients, which was worsened by the presence of DAN (Fig. 4A and online table

2). In addition, BM cells from T1D (STZ), T2D (Ob/Ob) and sympathectomised (6-OHDA)

mice showed increased p66Shc gene expression compared to controls (Fig. 4B). The

functional consequence of this association was tested using p66Shc-/-

mice which, once made

diabetic by STZ injection (T1D) or by crossing with Ob/Ob mice (T2D), were protected from

BM sympathectomy, as evidenced by the preserved number of Tyr-OH+ fibers (Fig. 4C, D).

As a result, T1D (STZ) and T2D (Ob/Ob) p66Shc-/-

mice were able to mobilize LKS cells and

EPC after G-CSF, despite they were as hyperglycaemic as wild type diabetic mice (Fig. 4E).

To confirm that restored mobilization in p66Shc-/-

diabetic mice was due to preserved BM

innervation, we show that STZ diabetic p66Shc-/-

mice treated with 6-OHDA, which

effectively denervates the BM despite p66Shc KO (Fig. 4D), were unable to mobilize LKS

cells after G-CSF stimulation (Fig. 4F).

Sirtuin-1 downregulation contributes to impaired stem cell mobilization in diabetes.

As sirtuin-1 (Sirt1) has been implicated in EPC survival and proangiogenic activity (30), we

analyzed its contribution in the mobilopathy model. We first found that Sirt1 gene expression

was reduced in PBMC of diabetic patients with DAN compared to those without (Fig. 5A and

online table 2). Consistently, Sirt1 gene expression was reduced in BM cells of T1D and T2D

and sympathectomised mice compared to controls (Fig. 5B). The link between neuropathy

and BM Sirt1 downregulation is strengthened by the evidence that preservation of BM

innervation by p66Shc deletion prevented BM Sirt1 mRNA reduction induced by T1D (STZ)

and T2D (Ob/Ob) (Fig. 5B). A cAMP-dependent activation of Sirt1 has been recently

Page 12 of 45Diabetes

Page 13: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

13

described (31) and we show here that treatment of human PBMC with the non-selective β-

adrenergic agonist isoproterenol stimulated Sirt1 mRNA expression in vitro (Online Fig. 3),

providing a link between sympathectomy and reduced BM Sirt1 expression. Consistently, we

also found that potentiation of NE signalling with desipramine restored Sirt1 gene expression

in the BM of T1D (STZ) mice (Fig. 5C).

To understand the functional consequences of Sirt1 downregulation in the BM, we generated

hematopoietic-restricted Sirt1-/-

mice (Vav1-Sirt1-/-

), lacking Sirt1 in cells that express the

Vav1 guanine nucleotide exchange factor, which is specific for the hematopoietic system

(Online Fig. 4) (32). Despite inconsistent data have been reported on hematopoietic function

in ubiquitous and hematopoietic Sirt1-/-

mice (33; 34), Vav1-Sirt1-/- mice, when subjected to

G-CSF stimulation and hind limb ischemia, recapitulated the poor mobilizer phenotype

observed in diabetic and sympathectomised mice (Fig. 5D), supporting that Sirt1

downregulation in the BM impairs mobilization. To confirm this hypothesis, we generated

Vav1-Sirt1 transgenic (TG) mice (Vav1-Sirt1TG

), which show a 12-fold increased expression

of Sirt1 in BM LKS cells (Online Fig. 4). Once made diabetic with STZ or sympathectomized

with 6-OHDA, Vav1-Sirt1TG

mice were able to mobilize LKS cells and EPC after G-CSF and

hind limb ischemia (Fig. 5E,F) and showed improved post-ischemic paw perfusion (Fig.

5G,H). To test the role of Vav1-expressing cells in the ischemic vasculature, we analyzed

Vav1-YFP mice, which express the yellow fluorescent protein only in hematopoietic (Vav1+)

cells. Based on the integration of YFP+ cells into the muscle vascular endothelium of ischemic

compared to non-ischemic hind limbs, we show that Vav1+ cells can contribute to post-

ischemic angiogenesis (Online Fig. 5), explaining why modulation of Sirt1 in Vav1+ cells can

translate into improved recovery after ischemia. These data indicate that reduced Sirt1

expression in the BM is responsible for the impaired stem cell mobilization in diabetic and

sympathectomised mice and that this can impact on vascular recovery.

Page 13 of 45 Diabetes

Page 14: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

14

Overexpression of adhesion molecules impair stem cell mobilization in diabetes and

neuropathy.

Data obtained in Vav1-Sirt1KO/TG

mice indicate that mobilization is affected by hematopoietic

cell-intrinsic mechanisms in diabetic and sympathectomised mice. In addition, the mechanism

of impaired stem cell mobilization associated with DAN are likely to be very downstream in

the cascade of events that make cells leaving the niche, as it is common to G-CSF and

ischemia, which act on different niche components (35). Therefore, we analyzed the gene

expression of a series of typical niche adhesion molecules (CD11a, CD11c, CD49d, CD49e,

CD62L, ICAM1) in BM cells and found they were globally upregulated in diabetic and

sympathectomised mice compared to controls (Online Fig. 6). The link between diabetic BM

neuropathy and changes in gene expression is supported by the finding that upregulation of

adhesion molecule genes occurred after the development of neuropathy in T1D mice (Online

Fig 7). We focused on CD62L/L-selectin because overexpression of this molecule was

previously noted by others in the diabetic BM (7). Upregulation of CD62L was confirmed by

flow cytometry on LKS cells of T1D (STZ), T2D (Ob/Ob), sympathectomised and Vav1-

Sirt1-/-

mice, while it was prevented in p66Shc-/- LKS cells of diabetic and sympathectomised

mice (Fig. 6A,B). The link between adrenergic signalling, Sirt1 and CD62L expression is

supported by the decrease in CD62L mRNA after treatment of PBMC with isoproterenol

(Online Fig. 1). L-selectin (CD62L) is prominently involved in the trafficking of blood cells

to lymphoid tissues (36), and excess L-selectin expression may render the cells more adherent

to the BM stroma, preventing mobilization. The relevance of L-selectin overexpression in this

setting is confirmed by the observation that genetic L-selectin deletion (Sell-/-

) partially

restored mobilization in diabetic and sympathectomised mice (Fig. 6C).

Page 14 of 45Diabetes

Page 15: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

15

DISCUSSION

This study shows for the first time that autonomic neuropathy in the BM impairs stem cell

mobilization in diabetes with dysregulation of the lifespan determinant genes p66Shc and

Sirt1. Previous data on BM SNS in diabetes models were inconsistent, reporting either

reduced (37) or increased (7) fibers. Our data indicate that T1D (STZ) and T2D (Ob/Ob) in

mice are characterized by a profound depletion of BM SNS fibers. We also show that BM

denervation in models of diabetes occurs via p66Shc. p66Shc is induced by hyperglycemia

(38) and mediates the development of several complications of experimental diabetes

including cardiomyopathy (39), nephropathy (40) and delayed wound healing (41). In

addition, p66Shc contributes to the hyperglycaemic damage of BM-derived EPC (42).

Although the link between DAN and mobilopathy cannot be firmly established, the

preservation of BM innervation and the rescue of mobilization in p66Shc-/-

diabetic mice

suggests that neuropathy impairs mobilization in models of diabetes. Other eventual effects of

p66Shc deletion seem important in this model, because chemical denervation abolished the

mobilization rescue in diabetic p66Shc-/-

mice. The mobilizing activity of G-CSF is dependent

on the SNS (14), and we show that also hind limb ischemia requires a functional SNS system

to elicit stem/progenitor mobilization, as it is defective in sympathectomised mice. On the

other hand, AMD3100 directly disrupts the CXCR4/CXCL12 interaction to promote

mobilization independently of the SNS (24). The effectiveness AMD3100 in inducing

mobilization in diabetic mice and the inefficacy of both SNS-dependent stimuli (G-CSF and

ischemia) is in line with the hypothesis that neuropathy impairs mobilization in diabetes. In

addition, potentiation of residual SNS output by treatment with the NE reuptake inhibitor

desipramine partially restored G-CSF-induced mobilization in diabetic mice. It has been

demonstrated that G-CSF stimulates the SNS by reducing NE reuptake, while desipramine

Page 15 of 45 Diabetes

Page 16: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

16

can rescue G-CSF-triggered mobilization in poor mobilizer mice (25). These data suggest that

a defective adrenergic signalling is responsible for impaired mobilization in diabetes and

suggests a potential treatment strategy. We have previously shown that T1D and T2D patients

have impaired G-CSF induced mobilization, irrespectively of neuropathy (9). However, SNS

fiber loss can be present in up to 50% of patients without clinical/instrumental evidence of

DAN (43). Thus, investigation into the history and features of diabetic BM dysautonomy is an

interesting matter of future research.

Sirtuin-1 (Sirt1) has been implicated in EPC survival and proangiogenic activity (30), its

expression is reduced in EPC of T2D patients (44), and is lowered by high glucose in THP-1

cells (45). Diabetic as well as chemically induced neuropathy reduced Sirt1 in the BM. The

mechanism whereby the SNS regulates Sirt1 expression remains unclear, but we show that

activation of the beta-adrenergic receptor increases Sirt1 mRNA in blood cells in vitro. The

link between neuropathy and Sirt1 repression is strengthened in vivo by the observation that

potentiating NE signalling with desipramine rapidly restored Sirt1 expression in the BM of

T1D mice. In addition, induction of diabetes in p66Shc-/-

was not followed by Sirt1 mRNA

reduction in the BM, which can be attributed to the preserved BM innervation, although we

cannot definitely rule out that metabolic dysregulations independent of neuropathy can affect

Sirt1 expression. Importantly, we show that Sirt1 downregulation in BM cells is causally

linked to impaired mobilization. In fact, hematopoietic-specific Sirt1 knock-out recapitulated

the poor mobilizer phenotype, while hematopoietic Sirt1 overexpression rescued mobilization

in diabetic and sympathectomised mice. As hematopoietic cell-intrinsic modulation of Sirt1

was sufficient to influence mobilization, we focused on a series of adhesion molecules

typically implicated in the trafficking of stem cells in the BM. The generalized induction of

adhesion molecule genes in diabetic and sympathectomized mice, by rendering the cells more

adherent to the stroma, can thus prevent mobilization. Excess CD62L (L-selectin) expression

Page 16 of 45Diabetes

Page 17: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

17

on BM LKS of diabetic mice was previously noted (7), and we now report that this is in fact

implicated in the diabetes- and neuropathy-induced mobilopathy, as L-selectin deletion was

sufficient to rescue mobilization in both models. Other adhesion molecules likely provide

redundant signals, such that moderate increases in their expression may additively promote

stem cell retention.

Taken together, our data suggest that BM autonomic neuropathy induced by diabetes via

p66Shc plays a central role in the impaired BM stem cell mobilization, involving

dysregulation of hematopoietic Sirt1 and cell-intrinsic mechanisms including adhesion

molecule overexpression. As multiple redundant mechanisms regulate the complex stem cell

niche, it is likely that SNS-independent pathways contribute to diabetes-induced impairment

in stem cell mobilization. Importantly, both in mice and in humans, diabetes induces

extensive microvascular remodelling of the BM (6). We now expand this notion, by showing

that neuropathy is an important component of diabetic niche remodelling, with negative

functional implications for mobilization.

Vascular stem/progenitor cells mobilized from the BM by G-CSF have been shown to

stimulate endothelial healing and angiogenesis (46; 47). In addition, ischemia-induced

mobilization is considered a natural response to tissue damage that promotes angiogenesis and

reperfusion (11). Therefore, impaired mobilization of cells with vasculotropic activity is

expected to promote cardiovascular disease. We show that the poor mobilizer phenotype of

diabetic and sympathectomised mice is associated with defective recovery after ischemia.

While several BM-independent mechanisms can contribute to this phenomenon, it is of

interest that cell-intrinsic Sirt1 overexpression rescues at the same time post-ischemic

mobilization and reperfusion. The relevance of Sirt1 modulation in Vav1-expressing cells is

supported by the contribution played by Vav1+ cells to the microvasculature of ischemic

Page 17 of 45 Diabetes

Page 18: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

18

muscles. Therefore, our data suggest that rescue of BM function in diabetes can promote

recovery from ischemia.

DAN is a disabling condition with reduced life expectancy and increased cardiovascular risk

(18). Our data suggest a novel possible mechanism whereby DAN worsens vascular health.

Future investigation into the features of BM neuropathy in humans and its relationships with

cardiovascular DAN will shed light on this novel diabetic complication. In addition, diabetes

is considered an accelerated aging disease (48) and dysregulation of longevity-determinant

hubs, such as Sirt1 and p66Shc, emerges as critical in inducing aging features including

autonomic neuropathy, impaired stem cell mobilization and defective recovery from ischemia.

Therefore, countering aging-associated pathways has the potential to tackle vascular

complications of diabetes, by restoring BM function and BM-derived stem/progenitor cells.

ACKNOWLEDGEMENTS

Author contribution: M.A. designed and performed in vivo and ex vivo experiments and

contributed to manuscript writing; N.P., S.C. and G.C. performed molecular biology analyses;

M.T. and R.M. generated and analyzed transgenic Vav1 mice; L.M., M.G. and P.P. generated

and analyzed p66Shc KO mice; S.V.dK. contributed to patient recruitment and

characterization; A.A. supervised the project, provided financial support and wrote the

manuscript; G.P.F. designed the project, recruited and characterized patients, analyzed data,

wrote the manuscript and provided financial support.

GPF is the guarantor of the study. The authors have no conflicting financial interests. This

study was supported by EFSD/Lilly Grant & Fellowship program to GPF, an Italian Ministry

of Health grant (GR-2010-2301676) to GPF, LOEWE CGT and SBF834 B8/Z1 grants to MT.

Page 18 of 45Diabetes

Page 19: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

19

REFERENCES

1. Fadini GP, Sartore S, Agostini C, Avogaro A: Significance of endothelial progenitor cells

in subjects with diabetes. Diabetes Care 30:1305-1313, 2007

2. Fadini GP, Boscaro E, de Kreutzenberg S, Agostini C, Seeger F, Dimmeler S, Zeiher A,

Tiengo A, Avogaro A: Time course and mechanisms of circulating progenitor cell reduction

in the natural history of type 2 diabetes. Diabetes Care 33:1097-1102, 2010

3. Fadini GP, Losordo D, Dimmeler S: Critical reevaluation of endothelial progenitor cell

phenotypes for therapeutic and diagnostic use. Circ Res 110:624-637, 2012

4. Oikawa A, Siragusa M, Quaini F, Mangialardi G, Katare RG, Caporali A, van Buul JD, van

Alphen FP, Graiani G, Spinetti G, Kraenkel N, Prezioso L, Emanueli C, Madeddu P: Diabetes

mellitus induces bone marrow microangiopathy. Arterioscler Thromb Vasc Biol 30:498-508,

2010

5. Mangialardi G, Katare R, Oikawa A, Meloni M, Reni C, Emanueli C, Madeddu P: Diabetes

Causes Bone Marrow Endothelial Barrier Dysfunction by Activation of the RhoA-Rho-

Associated Kinase Signaling Pathway. Arterioscler Thromb Vasc Biol 33:555-564, 2013

6. Spinetti G, Cordella D, Fortunato O, Sangalli E, Losa S, Gotti A, Carnelli F, Rosa F,

Riboldi S, Sessa F, Avolio E, Beltrami AP, Emanueli C, Madeddu P: Global remodeling of

the vascular stem cell niche in bone marrow of diabetic patients: implication of the

microRNA-155/FOXO3a signaling pathway. Circ Res 112:510-522, 2013

7. Ferraro F, Lymperi S, Mendez-Ferrer S, Saez B, Spencer JA, Yeap BY, Masselli E, Graiani

G, Prezioso L, Rizzini EL, Mangoni M, Rizzoli V, Sykes SM, Lin CP, Frenette PS, Quaini F,

Scadden DT: Diabetes impairs hematopoietic stem cell mobilization by altering niche

function. Sci Transl Med 3:104ra101, 2011

Page 19 of 45 Diabetes

Page 20: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

20

8. Fadini GP, Albiero M, Seeger F, Poncina N, Menegazzo L, Angelini A, Castellani C,

Thiene G, Agostini C, Cappellari R, Boscaro E, Zeiher A, Dimmeler S, Avogaro A: Stem cell

compartmentalization in diabetes and high cardiovascular risk reveals the role of DPP-4 in

diabetic stem cell mobilopathy. Basic Res Cardiol 108:313, 2013

9. Fadini GP, Albiero M, Vigili de Kreutzenberg S, Boscaro E, Cappellari R, Marescotti M,

Poncina N, Agostini C, Avogaro A: Diabetes Impairs Stem Cell and Proangiogenic Cell

Mobilization in Humans. Diabetes Care, 2012

10. Fadini GP, Avogaro A: Diabetes impairs mobilization of stem cells for the treatment of

cardiovascular disease: A meta-regression analysis. Int J Cardiol, 2012

11. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, Magner M, Isner JM,

Asahara T: Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial

progenitor cells for neovascularization. Nat Med 5:434-438, 1999

12. Tamarat R, Silvestre JS, Le Ricousse-Roussanne S, Barateau V, Lecomte-Raclet L,

Clergue M, Duriez M, Tobelem G, Levy BI: Impairment in ischemia-induced

neovascularization in diabetes: bone marrow mononuclear cell dysfunction and therapeutic

potential of placenta growth factor treatment. Am J Pathol 164:457-466, 2004

13. DiPersio JF: Diabetic stem-cell "mobilopathy". N Engl J Med 365:2536-2538, 2011

14. Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, Frenette PS:

Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from

bone marrow. Cell 124:407-421, 2006

15. Spiegel A, Shivtiel S, Kalinkovich A, Ludin A, Netzer N, Goichberg P, Azaria Y, Resnick

I, Hardan I, Ben-Hur H, Nagler A, Rubinstein M, Lapidot T: Catecholaminergic

neurotransmitters regulate migration and repopulation of immature human CD34+ cells

through Wnt signaling. Nat Immunol 8:1123-1131, 2007

Page 20 of 45Diabetes

Page 21: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

21

16. Dar A, Schajnovitz A, Lapid K, Kalinkovich A, Itkin T, Ludin A, Kao WM, Battista M,

Tesio M, Kollet O, Cohen NN, Margalit R, Buss EC, Baleux F, Oishi S, Fujii N, Larochelle

A, Dunbar CE, Broxmeyer HE, Frenette PS, Lapidot T: Rapid mobilization of hematopoietic

progenitors by AMD3100 and catecholamines is mediated by CXCR4-dependent SDF-1

release from bone marrow stromal cells. Leukemia 25:1286-1296, 2011

17. Lucas D, Scheiermann C, Chow A, Kunisaki Y, Bruns I, Barrick C, Tessarollo L, Frenette

PS: Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration.

Nat Med 19:695-703, 2013

18. Vinik AI, Ziegler D: Diabetic cardiovascular autonomic neuropathy. Circulation 115:387-

397, 2007

19. Hage FG, Iskandrian AE: Cardiac autonomic denervation in diabetes mellitus. Circ

Cardiovasc Imaging 4:79-81, 2011

20. Antonelli Incalzi R, Fuso L, Giordano A, Pitocco D, Maiolo C, Calcagni ML, Ghirlanda

G: Neuroadrenergic denervation of the lung in type I diabetes mellitus complicated by

autonomic neuropathy. Chest 121:443-451, 2002

21. Pozzoli O, Vella P, Iaffaldano G, Parente V, Devanna P, Lacovich M, Lamia CL, Fascio

U, Longoni D, Cotelli F, Capogrossi MC, Pesce M: Endothelial fate and angiogenic

properties of human CD34+ progenitor cells in zebrafish. Arterioscler Thromb Vasc Biol

31:1589-1597, 2011

22. Fadini GP, de Kreutzenberg S, Agostini C, Boscaro E, Tiengo A, Dimmeler S, Avogaro

A: Low CD34+ cell count and metabolic syndrome synergistically increase the risk of adverse

outcomes. Atherosclerosis 207:213-219, 2009

23. Weiss RB: Streptozocin: a review of its pharmacology, efficacy, and toxicity. Cancer

Treat Rep 66:427-438, 1982

Page 21 of 45 Diabetes

Page 22: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

22

24. Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper S, Plett PA, Liles WC, Li X,

Graham-Evans B, Campbell TB, Calandra G, Bridger G, Dale DC, Srour EF: Rapid

mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a

CXCR4 antagonist. J Exp Med 201:1307-1318, 2005

25. Lucas D, Bruns I, Battista M, Mendez-Ferrer S, Magnon C, Kunisaki Y, Frenette PS:

Norepinephrine reuptake inhibition promotes mobilization in mice: potential impact to rescue

low stem cell yields. Blood 119:3962-3965, 2012

26. Bento CF, Pereira P: Regulation of hypoxia-inducible factor 1 and the loss of the cellular

response to hypoxia in diabetes. Diabetologia 54:1946-1956, 2011

27. Waltenberger J: Impaired collateral vessel development in diabetes: potential cellular

mechanisms and therapeutic implications. Cardiovasc Res 49:554-560, 2001

28. Recalde A, Richart A, Guerin C, Cochain C, Zouggari Y, Yin KH, Vilar J, Drouet I, Levy

B, Varoquaux O, Silvestre JS: Sympathetic nervous system regulates bone marrow-derived

cell egress through endothelial nitric oxide synthase activation: role in postischemic tissue

remodeling. Arterioscler Thromb Vasc Biol 32:643-653, 2012

29. Paneni F, Volpe M, Luscher TF, Cosentino F: SIRT1, p66Shc, and Set7/9 in Vascular

Hyperglycemic Memory: Bringing All the Strands Together. Diabetes 62:1800-1807, 2013

30. Zhao T, Li J, Chen AF: MicroRNA-34a induces endothelial progenitor cell senescence

and impedes its angiogenesis via suppressing silent information regulator 1. Am J Physiol

Endocrinol Metab 299:E110-116, 2010

31. Gerhart-Hines Z, Dominy JE, Jr., Blattler SM, Jedrychowski MP, Banks AS, Lim JH,

Chim H, Gygi SP, Puigserver P: The cAMP/PKA pathway rapidly activates SIRT1 to

promote fatty acid oxidation independently of changes in NAD(+). Mol Cell 44:851-863,

2011

Page 22 of 45Diabetes

Page 23: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

23

32. Ogilvy S, Metcalf D, Gibson L, Bath ML, Harris AW, Adams JM: Promoter elements of

vav drive transgene expression in vivo throughout the hematopoietic compartment. Blood

94:1855-1863, 1999

33. Leko V, Varnum-Finney B, Li H, Gu Y, Flowers D, Nourigat C, Bernstein ID, Bedalov A:

SIRT1 is dispensable for function of hematopoietic stem cells in adult mice. Blood 119:1856-

1860, 2012

34. Singh SK, Williams CA, Klarmann K, Burkett SS, Keller JR, Oberdoerffer P: Sirt1

ablation promotes stress-induced loss of epigenetic and genomic hematopoietic stem and

progenitor cell maintenance. J Exp Med, 2013

35. Hoggatt J, Scadden DT: The stem cell niche: tissue physiology at a single cell level. J Clin

Invest 122:3029-3034, 2012

36. Ley K, Laudanna C, Cybulsky MI, Nourshargh S: Getting to the site of inflammation: the

leukocyte adhesion cascade updated. Nat Rev Immunol 7:678-689, 2007

37. Busik JV, Tikhonenko M, Bhatwadekar A, Opreanu M, Yakubova N, Caballero S, Player

D, Nakagawa T, Afzal A, Kielczewski J, Sochacki A, Hasty S, Li Calzi S, Kim S, Duclas SK,

Segal MS, Guberski DL, Esselman WJ, Boulton ME, Grant MB: Diabetic retinopathy is

associated with bone marrow neuropathy and a depressed peripheral clock. J Exp Med

206:2897-2906, 2009

38. Pagnin E, Fadini G, de Toni R, Tiengo A, Calo L, Avogaro A: Diabetes induces p66shc

gene expression in human peripheral blood mononuclear cells: relationship to oxidative stress.

J Clin Endocrinol Metab 90:1130-1136, 2005

39. Rota M, LeCapitaine N, Hosoda T, Boni A, De Angelis A, Padin-Iruegas ME, Esposito G,

Vitale S, Urbanek K, Casarsa C, Giorgio M, Luscher TF, Pelicci PG, Anversa P, Leri A,

Kajstura J: Diabetes promotes cardiac stem cell aging and heart failure, which are prevented

by deletion of the p66shc gene. Circ Res 99:42-52, 2006

Page 23 of 45 Diabetes

Page 24: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

24

40. Menini S, Amadio L, Oddi G, Ricci C, Pesce C, Pugliese F, Giorgio M, Migliaccio E,

Pelicci P, Iacobini C, Pugliese G: Deletion of p66Shc longevity gene protects against

experimental diabetic glomerulopathy by preventing diabetes-induced oxidative stress.

Diabetes 55:1642-1650, 2006

41. Fadini GP, Albiero M, Menegazzo L, Boscaro E, Pagnin E, Iori E, Cosma C, Lapolla A,

Pengo V, Stendardo M, Agostini C, Pelicci PG, Giorgio M, Avogaro A: The redox enzyme

p66Shc contributes to diabetes and ischemia-induced delay in cutaneous wound healing.

Diabetes 59:2306-2314, 2010

42. Di Stefano V, Cencioni C, Zaccagnini G, Magenta A, Capogrossi MC, Martelli F:

p66ShcA modulates oxidative stress and survival of endothelial progenitor cells in response to

high glucose. Cardiovasc Res 82:421-429, 2009

43. Scholte AJ, Schuijf JD, Delgado V, Kok JA, Bus MT, Maan AC, Stokkel MP,

Kharagitsingh AV, Dibbets-Schneider P, van der Wall EE, Bax JJ: Cardiac autonomic

neuropathy in patients with diabetes and no symptoms of coronary artery disease: comparison

of 123I-metaiodobenzylguanidine myocardial scintigraphy and heart rate variability. Eur J

Nucl Med Mol Imaging 37:1698-1705, 2010

44. Balestrieri ML, Servillo L, Esposito A, D'Onofrio N, Giovane A, Casale R, Barbieri M,

Paolisso P, Rizzo MR, Paolisso G, Marfella R: Poor glycaemic control in type 2 diabetes

patients reduces endothelial progenitor cell number by influencing SIRT1 signalling via

platelet-activating factor receptor activation. Diabetologia 56:162-172, 2013

45. de Kreutzenberg SV, Ceolotto G, Papparella I, Bortoluzzi A, Semplicini A, Dalla Man C,

Cobelli C, Fadini GP, Avogaro A: Downregulation of the longevity-associated protein sirtuin

1 in insulin resistance and metabolic syndrome: potential biochemical mechanisms. Diabetes

59:1006-1015, 2010

Page 24 of 45Diabetes

Page 25: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

25

46. Kong D, Melo LG, Gnecchi M, Zhang L, Mostoslavsky G, Liew CC, Pratt RE, Dzau VJ:

Cytokine-induced mobilization of circulating endothelial progenitor cells enhances repair of

injured arteries. Circulation 110:2039-2046, 2004

47. Minamino K, Adachi Y, Okigaki M, Ito H, Togawa Y, Fujita K, Tomita M, Suzuki Y,

Zhang Y, Iwasaki M, Nakano K, Koike Y, Matsubara H, Iwasaka T, Matsumura M, Ikehara

S: Macrophage colony-stimulating factor (M-CSF), as well as granulocyte colony-stimulating

factor (G-CSF), accelerates neovascularization. Stem Cells 23:347-354, 2005

48. Seshasai SR, Kaptoge S, Thompson A, Di Angelantonio E, Gao P, Sarwar N, Whincup

PH, Mukamal KJ, Gillum RF, Holme I, Njolstad I, Fletcher A, Nilsson P, Lewington S,

Collins R, Gudnason V, Thompson SG, Sattar N, Selvin E, Hu FB, Danesh J: Diabetes

mellitus, fasting glucose, and risk of cause-specific death. N Engl J Med 364:829-841, 2011

Page 25 of 45 Diabetes

Page 26: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

26

Table 1. Clinical characteristics of the study population. *p<0.05 DAN versus no DAN.

Characteristic No DAN DAN

Number 112 29

Age, years 59.1±11.1 52.2±13.8*

Sex male, % 66 59

Type 1 / type 2 diabetes 24/88 12/17*

Diabetes duration, years 10.5±9.9 12.0±10.2

HbA1c, % 7.7±1.5 8.8±2.1*

BMI, kg/m2 28.6±5.4 27.7±5.4

Waist circumference, cm 94.6±26.7 92.8±24.7

Smoking habit, % 14 14

Hypertension, % 61 66

Systolic blood pressure, mm Hg 131.7±16.8 130.2±18.5

Diastolic blood pressure, mm Hg 78.5±10.2 77.8±8.4

Heart rate, bpm 74.9±10.7 80.3±12.0*

Total cholesterol, mg/dl 181.9±41.3 179.4±56.1

HDL cholesterol, mg/dl 50.7±17.1 48.2±20.6

LDL cholesterol, mg/dl 108.9±28.8 110.3±35.8

Triglycerides, mg/dl 121.5±81.0 123.0±64.7

Coronary artery disease, % 7 10

Peripheral arterial disease, % 8 7

Cerebrovascular disease, % 38 45

Retinopathy, % 19 55*

Serum creatinine, umol/l 72.6±28.2 83.2±25.9

Albumin excretion rate, mg/g 63.7±274.6 167.5±358.5

Albumin excretion rate >30 mg/g, % 26.8 44.8

Insulin, % 38 57

Metformin, % 51 43

Sulphonylureas, % 17 11

Glinides, % 4 4

Glitazones, % 3 7

DPP-4 inhibitors, % 8 4

GLP-1 receptor agonists, % 1 0

ACEi / ARBs, % 63 54

Calcium antagonists, % 15 14

Beta-blockers, % 17 18

Aspirin, % 29 32

Statin, % 60 54

Fibrate, % 4 0

CD34+ / 106 cells 464±203 279±174

Page 26 of 45Diabetes

Page 27: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

27

FIGURE LEGENDS

Figure 1. Diabetic autonomic neuropathy reduces circulating stem cells. A-B) Circulating

CD34+ cell levels (expressed as cells/10

6 events in A or cells/mL in B) are progressively

reduced with increasing number of pathological DAN tests in diabetic patients. C-D) Diabetic

patients meeting diagnostic criteria for DAN had reduced CD34+ cells/10

6 events (C) and

CD34+ cells/mL (D), compared to DAN-free patients. E-F) A breakdown analysis for each

DAN test showed trend reductions of CD34+ cells. *p<0.05 pathologic vs normal test.

Figure 2. Experimental diabetes induces BM denervation. A) Representative staining of

BM sections for Tyr-OH (red), scale bar = 50 micron. B) Quantification of Tyr-OH staining

in non diabetic controls, T1D (STZ), T2D (Ob/Ob) and sympathectomised (6-OHDA)

animals (n=5/group). *p<0.05.

Figure 3. Impaired stem cell mobilization and ischemic response in diabetic and

sympathectomized mice. A) Mobilization of LKS cells and EPC after G-CSF in non

diabetic, T1D (STZ), T2D (Ob/Ob) and sympathectomised (6-OHDA) mice. Representative

FACS plots along with fold-change data are shown. *p<0.05 versus pre-G-CSF (n=5/group).

B) Mobilization of LKS cells and EPC at day 3 after hind limb ischemia in non diabetic, T1D

(STZ), T2D (Ob/Ob) and sympathectomised (6-OHDA) mice. Representative FACS plots

along with fold-change data are shown. *p<0.05 versus pre-ischemia (n=5/group). C)

Mobilization of LKS cell in non diabetic and STZ T1D mice after AMD3100 administration.

Representative FACS plots and fold-change data are shown. *p<0.05 versus pre-AMD3100

(n=5/group). D) LKS cell mobilization in T1D diabetic mice after G-CSF and G-CSF +

desipramine (Des). Representative FACS plots and fold-change data are shown. *p<0.05

Page 27 of 45 Diabetes

Page 28: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

28

versus baseline (n=3/group). E) Post-ischemic response of wild type non diabetic, T1D and

sympathectomised (6-OHDA) mice. Representative histological analyses of microvascular

density (red, isolectin B4; green, WGA) as well as ischemic / non ischemic ratio of the

capillary/fiber density are shown. Laser Doppler quantification of paw perfusion is also

reported. *p<0.05 versus non diabetic control (n=5/group).

Figure 4. p66Shc and DAN-associated mobilopathy. A) p66Shc gene expression in PBMC

of non diabetic individuals and diabetic patients with and without DAN. *p<0.05. B) Gene

expression of p66Shc in the BM of control, T1D (STZ) and sympathectomised (6-OHDA)

mice. *p<0.05 versus non diabetic controls (n=5/group). C) Quantification of Tyr-OH

immunoreactive sympathetic fibers in wild type and p66Shc-/-

T1D and T2D mice. *p<0.05

(n=5/group). D) Representative immunofluorescence images of Tyr-OH staining in control

and p66Shc-/-

T1D (STZ), T2D (Ob/Ob) and sympathectomised mice. E) LKS and EPC

mobilization in wild type and p66Shc-/-

T1D (STZ) and T2D (Ob/Ob) mice after G-CSF.

*p<0.05 versus baseline (n=5/group). F) LKS and EPC mobilization in wild type T1D (STZ)

mice, p66Shc-/-

T1D mice and p66Shc-/-

T1D 6-OHDA treated mice after G-CSF. *p<0.05

versus baseline (n=3-5/group).

Figure 5. Sirt1 regulates stem cell mobilization and post-ischemic response. A) Sirt1

expression in PBMC of diabetic patients with and without DAN. B) Sirt1 expression in the

BM of wild type and p66Shc-/-

non diabetic control (CTRL), T1D (STZ), T2D (Ob/Ob) and

sympathectomised (6-OHDA) mice. *p<0.05 versus controls (n=5/group). C) BM Sirt1 gene

expression in non diabetic controls (CTRL), T1D (STZ) mice and T1D (STZ) mice treated

with desipramine. *p<0.05 versus control. D) LKS cell and EPC mobilization in

hematopoietic restricted (Vav1-)Sirt1-/-

mice after G-CSF (red) and hind limb ischemia (blue).

Page 28 of 45Diabetes

Page 29: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

29

*p<0.05 post vs pre (n=5/group). E) LKS cell and EPC mobilization in diabetic (STZ)

hematopoietic restricted (Vav1)-Sirt1 overexpressing (TG) mice after G-CSF (red) and hind

limb ischemia (blue). *p<0.05 post vs pre (n=5/group). F) LKS cell and EPC mobilization in

sympathectomised (6-OHDA) hematopoietic restricted Vav1-Sirt1TG

mice after G-CSF (red)

and hind limb ischemia (blue). *p<0.05 post vs pre (n=5/group). G) Post-ischemic response of

diabetic and sympathectomised Vav1-Sirt1TG

mice compared to the respective wild type

controls. Representative histological analyses of the microvasculature (red, isolectin B4;

green, laminin), as well as Laser Doppler perfusion (F) are shown. *p<0.05 versus wild type

normal controls (n=5/group).

Figure 6. CD62L (L-selectin) and stem cell mobilization. A) BM expression of L-selectin

(Sell) in wild type, p66Shc-/-

and Vav1-Sirt1TG

non diabetic, T1D (STZ) and

sympathectomized (6-OHDA) mice. *p<0.05 versus non diabetic. B) Mean fluorescence

intensity (MFI) of surface CD62L expression by flow cytometry on wild type, p66Shc-/-

and

Vav1-Sirt1-/-

mice. *p<0.05 versus non diabetic controls. C) LKS cell and EPC mobilization

in L-selectin KO (Sell-/-

) T1D and sympathectomised mice. *p<0.05 versus baseline

(n=5/group).

Page 29 of 45 Diabetes

Page 30: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

0 1 2 3 40

500

1000

1500

Number of altered DAN tests

CD

34

+ c

ell

s /

10

6

0 1 2 3 40

2000

4000

6000

8000

10000

12000

Number of altered DAN tests

CD

34

+ c

ell

s /

mL

No DAN DAN0

500

1000

1500

CD

34

+ c

ell

s /

10

6

No DAN DAN0

2000

4000

6000

8000

10000

12000

CD

34

+ c

ell

s /

mL

Dee

p Bre

athin

g

Orthost

atic

Hyp

otensi

on

Lying

-to-s

tandin

g

Val

salv

a m

aneu

vre

0

200

400

600Normal

Pathologic

CD

34

+ c

ell

s /

10

6

* **

p=0.07

Dee

p Bre

athin

g

Orthost

atic

Hyp

otensi

on

Lying

-to-s

tand

ing

Val

salv

a m

aneu

vre

0

1000

2000

3000

4000Normal

PathologicC

D34

+ c

ell

s /

mL

*

p=0.08

p<0.001 p<0.001

A

C

E

B

D

F

Figure 1

Page 30 of 45Diabetes

Page 31: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

Figure 2

Non diabetic T1D (STZ) T2D (Ob/Ob) 6-OHDA0

1

2

3

4

5

Ty

r-O

H+ f

ibers

/ H

PF

* **

A

B

T1D (STZ)

T2D (Ob/Ob) 6-OHDA

Non diabetic

Page 31 of 45 Diabetes

Page 32: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

pre-GCSF

post-GCSF

pre-GCSF

post-GCSF

pre-ischemia

14d post-

ischemia

pre-ischemia

3d post-

ischemia

pre-AMD3100

4h after

AMD3100

4-day G-CSF Hind limb ischemia

AMD3100 (Plerixafor) Desipramine + G-CSF

Non diabetic T1D (STZ) 6-OHDA0.0

0.5

1.0

1.5

Isch

em

ic / n

on

isch

em

ic r

ati

o

p=0.08

Non diabetic T1D (STZ) 6-OHDA0.0

0.2

0.4

0.6

Isch

em

ic / n

on

isch

em

ic r

ati

o

*

*

Non diabetic T1D (STZ) 6-OHDA

No

n i

sc

he

mic

Isch

em

icL

ase

r D

op

ple

r p

erf

usio

n

Non diabetic T1D (STZ) 6-OHDA

Isolectin WGA

Figure 3A B

C D

E

Page 32 of 45Diabetes

Page 33: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

*

Tyr-

OH

+fi

bers

/ H

PF

Wild type non

diabeticWild type

T1D (STZ)

p66Shc-/-

T1D (STZ)

Wild type

T2D (Ob/Ob)

p66Shc-/-

T2D (Ob/Ob)

**

*

non DM DM no DAN DM DAN0

1

2

3

4

p66S

hc /ββ ββ

-acti

n r

ati

o

*

*

Non diabetic T1D (STZ) T2D (Ob/Ob) 6-OHDA

0.5

1.0

1.5

2.0

* * *

BM

p66S

hc m

RN

A e

xp

ress

ion

Pre-GCSF Post-GCSFPre-GCSF Post-GCSF

*

*

*

*

*

*

T1D (STZ) T2D (Ob/Ob) 6-OHDA

Wild

-typ

ep66Shc-/-

Figure 4A B

C D

FE

Page 33 of 45 Diabetes

Page 34: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

CTR

L

T1D (S

TZ)

T2D (O

b/Ob)

6-OHDA

0.0

0.5

1.0

1.5

2.0

2.5WIld type

p66Shc-/-

BM

Sir

t1 e

xp

ressio

n

Figure 5

No DAN DAN0.0

0.5

1.0

1.5

2.0

Sir

t1 /ββ ββ

-acti

n r

ati

o

p<0.005A B

**

*

C

*

D E F4 days G-CSF Hind limb ischemia 4 days G-CSF Hind limb ischemia 4 days G-CSF Hind limb ischemia

CTR

LT1D

T1D +

Des

0.0

0.5

1.0

1.5

BM

Sir

t1 e

xp

ressio

n

G H

0.0

0.5

1.0

1.5

Non diabetic T1D (STZ) 6-OHDA

Isch

em

ic / n

on

isch

em

ic r

ati

oVav1-Sirt1TG T1D (STZ) Vav1-Sirt1TG 6-OHDA

No

n Is

ch

em

icIs

ch

em

ic

Vav1-Sirt1TG T1D (STZ) Vav1-Sirt1TG 6-OHDA

Laser

Do

pp

ler

perf

usio

n

0.0

0.2

0.4

0.6

Non diabetic T1D (STZ) 6-OHDA

Isch

em

ic / n

on

isch

em

ic r

ati

o

n.s.

*

*n.s.

Wild type Vav1-Sirt1TG Wild type Vav1-Sirt1TG

*

*

*

*

*

*

*

*

*

*

*

*

Page 34 of 45Diabetes

Page 35: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

Figure 6

Non d

iabet

ic

T1D (S

TZ)

6-OHDA

Non d

iabet

ic

T1D (S

TZ)

6-OHDA

Non d

iabet

ic

T1D (S

TZ)

6-OHDA

0

1

2

3

CD

62L

MF

I

p66Shc-/-

CD62L-PerCP-Cy5.5

Co

un

ts

Isotype

WT Non diabetic

WT T1D (STZ)

Isotype

p66Shc-/- T1D (STZ)

WT T1D (STZ)

Isotype

WT Non diabetic

Vav1-Sirt1KO

Vav1-Sirt1-/-

*

*

*

Wild-type

n.s.

p66Shc-/- Vav1-Sirt1TG

Non d

iabet

ic

T1D (S

TZ)

6-O

HDA

Non d

iabet

ic

T1D (S

TZ)

6-O

HDA

Non d

iabet

ic

T1D (S

TZ)

6-OHDA

0

1

2

3

4

Sell

BM

exp

ressio

n

Wild-type

* *

n.s.

n.s.

A B

C

*

* *

Page 35 of 45 Diabetes

Page 36: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

1

Albiero et al.

Diabetes causes bone marrow autonomic neuropathy

and impairs stem cell mobilization via dysregulated p66Shc and Sirt1

ONLINE SUPPLEMENTAL MATERIAL

Online Table 1. Multivariable analysis with circulating CD34+ cell level as dependent variables and

clinical characteristics identified by univariate analysis as explanatory variables.

Variable Beta-coefficient p-value

DAN -0.377 <0.001

Age -0.101 0.297

Diabetes type 0.151 0.121

HbA1c -0.141 0.085

Retinopathy 0.096 0.289

Heart rate 0.023 0.791

Page 36 of 45Diabetes

Page 37: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

2

Online Table 2. Clinical characteristics of patients with available measure of PBMC expression of p66Shc and Sirt1 (extracted from table 1).

Characteristic No DAN DAN

Age, years 60.0±14.0 59.7±15.0

Sex male, % 70 70

Type 1 / type 2 diabetes 3/7 3/7

Disease duration, years 10.0±7.8 12.9±7.8

HbA1c, % 7.7±1.8 7.9±1.4

p66Shc / β-actin expression 1.01±0.15 1.63±0.95

Sirt1 / β-actin expression 1.09±0.29 0.75±0.17

Page 37 of 45 Diabetes

Page 38: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

3

Online Table 3. Primer sequences.

Genes FW primer sequence RV primer sequence

Sequence accession

number

Mouse genes

vascular cell adhesion molecule 1 (Vcam1) TATGTCAACGTTGCCCCCAA GCTGTCTGCTCCACAGGATT NM_011693.3

src homology 2 domain-containing

transforming protein C1 (Shc1) – p66shc TGACAGGATGGCTGGCTT ACGGACTTCATGGTCTCC NM_001113331.2

intercellular adhesion molecule 1 (Icam1) AGCTCGGAGGATCACAAACG TCCAGCCGAGGACCATACAG NM_010493.2

integrin alpha L (Itgal) – CD11a ACTTCCACTTCCCGATCTGC CCACCTTTTGGTCCCTTGGT NM_001253872.1

integrin alpha 4 (Itga4) – CD49d GTTCTCCACCAAGAGCGCAT GATGAGCCAGCGCTTCGAC NM_010576.3

integrin alpha 5 (Itga5) – CD49e GAACCCTGTGTCCTGCATCA TTGGAGTTCCACCTCGAAGC NM_010577.3

selectin, lymphocyte (Sell) – CD62L TGATGCAGGGTATTACGGGC CACTGGACCACTTGGCAGAT NM_001164059.1

integrin alpha X (Itgax) – CD11c TCTTCTGCTGTTGGGGTTTGT GAGCACACTGTGTCCGAACT NM_021334.2

Sirtuin 1 (Sirt1) CAGTAGCACTAATTCCAAGTTCTA TTGGCATATTCACCACCTAGC NM_001159589.1

ubiquitin C (Ubc) GCCCAGTGTTACCACCAAGA CCCATCACACCCAAGAACA NM_019639.4

Human Genes

sirtuin 1 (SIRT1) TACCGAGATAACCTTCTGTTCG GTTCGAGGATCTGTGCCAAT NM_012238.4

selectin L (SELL) – CD62L GCCCTCTGTTACACAGCTTCT GGCCCATAGTACCCCACATC NM_000655.4

actin, beta (ACTB) GGATGCCACAGGACTCCA AGAGCTACGAGCTGCCTGAC NM_001101.3

src homology 2 domain-containing

transforming protein C1 (SHC1) – p66shc AATCAGAGAGCCTGCCACATT CTCTTCCTCCTCCTCATC NM_001130040

Page 38 of 45Diabetes

Page 39: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

4

Online Figure 1. Baseline levels of EPC and LKS cells in diabetic and sympathectomized (6-OHDA) mice. * p<0.05 versus non-diabetic control mice. “n.s.” stands for “not significant” compared with

non-diabetic control mice.

Page 39 of 45 Diabetes

Page 40: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

5

Online Figure 2. Effects of STZ and 6-OHDA in vitro on apoptosis and necrosis of total murine bone marrow cells (A), murine CD34+ bone marrow cells (B) and growth of hematopoietic colonies from

murine bone marrow cells (C). In panel (A), *p<0.05 versus percentages in the control (CTRL)

condition. In (C), both STZ and 6-OHDA were incubated at 10 microM concentration.

A B

C

Page 40 of 45Diabetes

Page 41: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

6

Online Figure 3. Effects of isoproterenol on PBMC Sirt1 and CD62L expression. PBMC of healthy

human donors were incubated without or with isoproterenol 100 µM and gene expression of Sirt1 and

CD62L (L-selectin, Sell), were analyzed. The cellular concentration of cyclic AMP (cAMP) were also

determined to select isoproterenol concentrations that result in increased cAMP production. *p<0.05

100 vs 0 µM isoproterenol.

0 µµµµM 100 µµµµM0.0

0.5

1.0

1.5

2.0

[Isoproterenol]

Sirt1 m

RNA (fold change)

0 µµµµM 100 µµµµM0.0

0.5

1.0

1.5

2.0

[Isoproterenol]

CD62L m

RNA (fold change)

*

*

0 µµµµM 100 µµµµM0.0

0.5

1.0

1.5

2.0

[Isoproterenol]

cAMP (pg/m

L)

*

Page 41 of 45 Diabetes

Page 42: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

7

Online Figure 4. Generation and characterization of transgenic animals. The breeding strategy used to generate Vav1-Sirt1-/- and Vav1-Sirt1TG mice is shown. Knock-out and overexpression of Sirt1,

respectively, was confirmed by real time PCR on LKS cells isolated by FACS.

Vav1Cre/+:

Sirt1ex4∆flox/+

Sirt1ex4flox/floxVav1Cre/+:

Sirt1ex4∆flox/+

Sirt1ex4flox/flox

With DapiWithout Dapi

LKS sorting:

efficient knock-down /

overexpression

Lin–Lin–

cKit

+

Sca1+

Vav1Cre/+ Sirt1stop-floxVav1Cre/+ Sirt1stop-flox

Sirt1 expression

CtrKOOver

Page 42 of 45Diabetes

Page 43: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

8

Online Figure 5. Contribution of Vav1+ cells to the microvasculature of ischemic muscles. Vav1-YFP

mice were subjected to hind limb ischemia and muscle sections stained for Isolectin B4 (red) to

visualize the vascular network (nuclei counterstained in blue with Hoechst); the green/yellow signal

represents the spontaneous YFP fluorescence. Merged figures are shown. YFP-expressing cells, indicating Vav1+ cells were only found in sections of ischemic muscles and not of non ischemic

contralateral control muscles. Some YFP+ cells were clearly integrated into the vasculature co-staining

with Isolectin, while other YFP+ cells did not co-localize with the red Isolectin signal and were

considered not integrated (likely intravascular). The right panel shows quantification of total

(integrated and non integrated) and integrated cells in ischemic and non ischemic muscle sections.

Hoechst

Isolectin

YFP

Ischemic muscle section

integrated

not

integrated

Non-ischemic muscle section

cells

+

Total YFP

cells

+

Integrated YFP

0

1

2

3

4

Ischemic muscle sections

Non-ischemic muscle sections

2.1

1.4

0.0 0.0

Cells / field

Page 43 of 45 Diabetes

Page 44: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

9

Online Figure 6. Expression of niche adhesion molecules in the BM. mRNA was extracted from BM cells of control non diabetic, T1D (STZ) and sympathectomised (6-OHDA) mice and analyzed for

expression of typical niche genes encoding for adhesion molecules (n=5/group). *p<0.05 versus non

diabetic control.

CD11a

CD11c

CD49d

CD49e

CD62L

ICAM1

0

1

2

3

4 CTRL

T1D (STZ)

6-OHDA

BM m

RNA expression

* *

* *

* *

* * *

*

Page 44 of 45Diabetes

Page 45: Page 1 of 45 Diabetes · EPC and LKS cells after ischemia, EPC levels peaked at day 3 after ischemia and LKS peaked at day 14. Therefore, these time points were used for all subsequent

10

Online Figure 7. Time course analysis of bone marrow neuropathy development and upregulation of adhesion molecules in T1D mice from 1 to 4 weeks after STZ administration. In the lower panel,

*p<0.05 versus basal; **p<0.05 versus 1 week.

CD49e

CD49d

ICAM1

CD62L

CD11a

CD11c

CD49e

CD49d

ICAM1

CD62L

CD11a

CD11c

CD49e

CD49d

ICAM1

CD62L

CD11a

CD11c

CD49e

CD49d

ICAM1

CD62L

CD11a

CD11c

CD49e

CD49d

ICAM1

CD62L

CD11a

CD11c

0

1

2

3

4

2 Week

3 Week

1 Week

Basal

4 Week

mRNA

(fold change vs basal) p<0.05 vs basal p<0.05 vs basal

n.s.

Page 45 of 45 Diabetes