• No results found

Characterisation of Torin-1 effects on human endothelial colony forming cells (ECFCs) biology

N/A
N/A
Protected

Academic year: 2021

Share "Characterisation of Torin-1 effects on human endothelial colony forming cells (ECFCs) biology"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Characterisation of Torin-1 effects on human endothelial colony forming cells (ECFCs) biology

L. E.R. Dietz

October 2016

Medina’s lab, Center for Experimental Medicine Queen’s University Belfast

Supervised by Dr. Reinhold J. Medina

Rijksuniversiteit Groningen Student number: S2250853 Supervisor: Dr. Marco C. Harmsen

Abstract

Diabetic Retinopathy (DR) is the most common microvascular complication of diabetes and the leading cause of blindness in working-aged adults. The hyperglycaemic milieu of diabetes causes functional impairment and loss of endothelial cells resulting in endothelial dysfunction. Endothelial progenitor cells (EPCs) have been recognised as playing a major role in vascular repair of ischaemic tissues as they are able to differentiate into mature endothelium. Endothelial colony forming cells (ECFCs), an EPC subtype, are considered as the bona fide EPCs. ECFCs are able to repair damaged vessels and therefore have a therapeutic potential in the treatment of DR. For clinical use, it is important that ECFCs can be expanded efficiently in vitro without the cells becoming senescent and losing proliferating potential. Although ECFCs can be efficiently expanded in vitro, their replicative potential is limited and the cells ultimately become senescent after long term culture.

mTOR inhibitors such as Rapamycin are found to be able to prolong lifespan. Torin-1, an mTORC1/2 inhibitor, was shown to suppress senescence more efficiently than rapamycin and therefore we focussed on Torin-1.

Our results provide the first characterisation of the effect of Torin-1 on ECFCs. We established that adding Torin-1 to healthy ECFCs is not beneficial for either proliferation or senescence

establishment. However, the drug seems to reduce the numbers of Etoposide-induced senescent ECFCs. Torin-1 is reported to activate autophagy. The complex relations between autophagy, senescence and apoptosis needs to be further elucidated. Furthermore, mTOR inhibition with Torin-1 significantly decreased mitochondrial respiration of ECFCs. Our study warrants further investigation in mTOR inhibitors and senescence and we speculate on the effects of mTOR inhibition on diabetic ECFCs.

(2)

Table of contents

Introduction……….. p. 3

Materials & Methods……….. p. 7

Results……… p. 10 Torin-1 has a cytostatic effect on proliferating ECFCs p. 10

Torin-1 and ECFCs senescence p. 12

Torin-1 effect on stress-induced senescent ECFCs p. 13 Torin-1 effectively activates autophagy molecular machinery in ECFCs p. 14 Torin-1 reduces oxygen consumption in ECFCs p. 16 Discussion……….……….………. p. 18

Conclusion……….……….……… p. 20

Further directions……….……….……… p. 20

References……….……….……… p. 21

Supplementary figures………... p. 25

(3)

Introduction

Diabetic Retinopathy (DR) is the most common microvascular complication of diabetes and among the leading causes of blindness in working age adults (Williams et al., 2004). DR can be classified into two clinical stages: non-proliferative and proliferative diabetic retinopathy (PDR). The first, non- proliferative stage is characterised by abnormal permeability, diminished capillary perfusion of retinal tissue, and formation of microaneurysms (Engerman, 1989). The abnormal capillary permeability can also cause macular oedema (MO), in which fluids and solutes leak into the surrounding retinal tissue, which collects around the macula. PDR develops as a result of the

ineffective re-vascularisation of the neuropile; the ischaemia caused by the breakdown of the retinal vasculature triggers the growth of new blood vessels on the surface of the retina towards the vitreous. These vessels are prone to leakage (He et al., 2015; Mohamed, Gillies, & Wong, 2007). The preretinal neovascularisation and MO may seriously impair vision, and ultimately lead to blindness.

Yet the pathogenesis of DR has not been fully elucidated. DR is considered a vascular disease as an effect of several diabetic factors, including the continuous hyperglycaemic condition characteristic of both type I and type II diabetes. The hyperglycaemic milieu causes functional impairment and loss of endothelial cells (ECs) resulting in endothelial dysfunction (Roberts & Porter, 2013).

The sustained hyperglycaemic environment and the resulting hypoxic conditions are known to upregulate various cytokines, including Vascular Endothelial Growth Factor (VEGF) (Boyer, Hopkins, Sorof, & Ehrlich, 2013; Dor, Porat, & Keshet, 2001). The overexpression of VEGF in DR promotes pathological angiogenesis and elevated vascular permeability, leading to diabetic MO and

contributing to the loss of visual function (Boyer et al., 2013; Stewart, Hayakawa, Akers, & Vinters, 1992). Currently, anti-VEGF agents are amongst the most common treatments for PDR, suppressing the pathological angiogenesis and bringing a halt to the progression of disease (Ajlan, Silva, & Sun, 2016). Still there is not a cure for DR. If the reparative angiogenesis in the retina could be targeted successfully, there is a possibility that it could restore endothelial function and eliminate the ischaemia that plays a major role in DR progression.

Bone-marrow-derived endothelial progenitor cells (EPCs) have been recognised as playing a major role in vascular repair of ischaemic tissues as they are able to differentiate into mature endothelium (Urbich & Dimmeler, 2004). Increasing the number of these cells might be an attractive therapeutic tool. EPCs can be isolated from peripheral or umbilical cord blood by in vitro culture and this

consistently produces two distinct EPC subtypes. Circulating angiogenic cells (CACs, previously named early EPCs) appear early in culture after 1-2 weeks, while Endothelial Colony Forming Cells (ECFCs, also known as OECs or late EPCs) appear later at 4-6 weeks (Hirschi, Ingram, & Yoder, 2008).

These EPC subtypes contribute to angiogenesis through different mechanisms. ECFCs can directly incorporate into resident vasculature, while CACs stimulate angiogenesis in a paracrine manner (Hur et al., 2004). ECFCs are considered the bona fide EPC and the most suitable EPC type for regenerative angiogenesis (Critser & Yoder, 2010; Lin, Weisdorf, Solovey, & Hebbel, 2000). Figure 1, taken from Medina et al 2013 in Stem Cells, shows a characterisation of ECFCs.

(4)

Previous studies have shown the in vitro and in vivo angiogenic potential of ECFCs; the ECFCs are capable of de novo tube formation and can repair vasculature by physically incorporating into newly formed blood vessels (Reinisch et al., 2009; Yoder et al., 2007). These studies showed that ECFCs have therapeutic potential, and our group has recently demonstrated that the ECFCs significantly increase the amount of normal vasculature and decrease avascular areas (Fig. 2, taken from Medina et al 2010 in Investigative ophthalmology & visual science).

Recent data from our lab demonstrate that although ECFCs can be efficiently expanded in vitro, their replicative potential is limited (Medina et al., 2013). Figure 3A shows that when ECFCs are amplified long-term in vitro, cell numbers reach a plateau of growth. This growth arrest was characterised as replicative senescence. These late passage cells acquired a larger and flatter morphology, typical of cellular senescence (Fig. 3B). The late passage ECFCs have increased senescence-associated beta-gal staining, a decrease in DNA replication, and an increase of DNA damage, as shown respectively by figure 3C, D and E. Together this indicates that after multiple cell divisions in vitro, ECFCs underwent replicative cellular senescence. Our lab showed that senescent ECFCs have significantly lowered proliferative, migratory and tube forming potential when compared to early passage cells (Medina et al., 2013). More importantly, the in vivo angiogenic capability of senescent ECFCs is also notably impaired. The senescence programme is of great importance for novel cell therapies that require

Figure 1. Characterisation of endothelial progenitor cells as the endothelial colony forming cell (ECFC) population. (A) ECFCs grow in culture as a cobblestone-shaped cell monolayer. Scale bar = 100 μm. (B) Immunofluorescence staining for Vimentin (red) and cadherin (green). Nuclei are counterstained with 4ʹ,6-diamidino-2-phenylindole (DAPI) (blue). (C) VEGFR2 expression is shown in green. (D) Caveolin 1 expression in ECFCs is shown in green. Scale bars in (B–D) = 5μm. (E) Immunophenotypic characterisation of ECFCs by flow cytometry. Cell surface antibodies are shown in red histograms, respective isotype controls in blue histograms. The percentage of positive cells appears in the top right of each panel.

(Medina et al., 2013)

Figure 2. ECFCs contribute to vascular repair of ischemic retina. (A, B) Representative flat mounted retinas of C57BL/6 mice injected with vehicle or ECFCs (OECs), respectively. Lectin staining (green) identifies retinal vasculature. Avascular regions are surrounded by a yellow line. Insets, white: avascular (ischemic) areas. Scale bars, 1 mm. (Medina et al., 2010).

A B

(5)

sufficient numbers of ECFCs. Therefore, optimisation of the ECFC number amplification process as well as prevention of cellular senescence is of great interest for effective translation into clinics.

Recently, interest has grown surrounding mTOR (mammalian Target Of Rapamycin) inhibitors due to multiple reports about their ability to prolong lifespan (Chen, Liu, Liu, & Zheng, 2009; Harrison et al., 2009a; Zhang et al., 2014). mTOR is suggested as a driver of conversion from reversible cell cycle arrest to senescence (geroconversion) and inhibitors such as rapamycin have been reported to suppress or decelerate geroconversion (Dulic, 2013; Leontieva & Blagosklonny, 2010; Luo et al., 2014). Rapamycin is an allosteric and indirect mTOR inhibitor, which in most conditions inhibits mTOR complex 1 (mTORC1) and not mTOR complex 2 (mTORC2) (Kang et al., 2013). In contrast, Torin-1 is an ATP-competitive direct inhibitor of the TOR kinase and is not selective; it abrogates all activities of both mTORC1 and mTORC2 (a “dual” mTOR inhibitor) (Benjamin et al., 2011; Yu et al., 2009). Leontieva and colleagues demonstrated that, at low concentrations, Torin-1 is capable of geroconversion (Leontieva et al., 2015). Torin-1 preserved the proliferative potential in growth arrested HT-p21 cells and fibroblasts, while it had a cytostatic effect on healthy proliferating cells.

The dual mTORC1/C2 inhibitor also prevented senescent morphology and hypertrophy, more than the conventional mTOR inhibitor rapamycin.

To test the effect of Torin-1 on geroconversion, an experimental model to induce senescence isneeded. Because replicative senescence is a time-consuming process, most researches use

B

C

E

Figure 3. In vitro expansion of ECFCs leads to the cells going into senescence. (A) Growth curves for ex vivo expanded cord blood-derived ECFCs (CB.OECs, green), peripheral blood-derived ECFCs (PB.OECs, red), human aortic endothelial cells (HAECs) and human dermal microvascular endothelial cells (HDMECs) in blue. (B): Representative phase-contrast micrographs of cultured ECFCs at different stages during their in vitro lifespan. (C) Representative images for early- and late-passage OECs stained for SA-β-Gal shown in blue. (D) Immunofluorescence staining for BrdU shown in green, nuclei are counterstained in red with PI. (E) Representative immunofluorescence staining for DNA damage marker γ-H2AX in green. Nuclei are counterstained in red with PI. Scale bars (B,C,D,E) = 100 μm. Taken from the journal Stem Cells (Medina et al., 2013)

D

C B

A

E

B

(6)

chemicals as an acute stress-induced senescence model. A frequently used senescence-inducing agent is Etoposide. The model for Etoposide stress-induced senescence is shown in figure 4 (taken from unpublished data from Michael McKee, former member of our group).

Currently there are no reports studying mTOR inhibitors on ECFCs. In the present study, we explored the effect of Torin-1 on the biology of ECFCs. We found that Torin-1 has a cytostatic and senescence- inducing effect on proliferating ECFCs, while it diminishes the number of senescent cells in the Etoposide model. We show that Torin-1 activates autophagy and decreases mitochondrial respiration in ECFCs and consider the implications of these findings for further research.

Figure 4. Acute, Stress-Induced Senescence in Etoposide Treated ECFCs. ECFCs underwent 24 hours of Etop Treatment.

These cells were counted and then re-plated at 1x105 cells (red horizontal line). (A) Following 4 further days of treatment the OECs were counted again (left y-axis) and the single cell suspension diameter was also recorded (right y-axis) after a total of 5 days treatment. OEC number and diameter were determined using a CASY cell counter. (B) OECs were incubated for 10 days in varying concentrations of Etoposide and stained for senescence associated (SA) β-Galactosidase (blue staining), which is a marker of cellular senescence. Images were then taken using a phase contrast microscope at 20x magnification. The summary figure (C) shows the percentage of β-Galactosidase stained cells in the treated and control groups. Etoposide treatment has been shown to inhibit proliferation of OECs and increase their average cell diameter (p<0.01). OECs treated with Etoposide exhibit blue staining when stained for SA β-Galactosidase (p<0.001 compared to controls), which is a marker of cellular senescence. Taken from thesis Michael McKee, 2014.

(7)

Materials and methods

Cell isolation and culture

The ECFCs were isolated from human umbilical cord blood (CB) and peripheral blood (PB) under full ethical approval and in accordance with the declaration of Helsinki . Mononuclear cells (MNCs) were isolated from PB by density gradient centrifugation. To obtain ECFCs, MNCs were seeded onto collagen coated wells at a density of 10x107 cells/ml using complete EGM-2 that contained fetal bovine serum (FBS, Gibco), VEGF, hydrocortisone, hFGF, R3-IGF-1, ascorbic acid, hEGF, Heparin, primocin (Invivogen, San Diego, US) and supplemented with 10% FBS (Lonza Ltd., Slough, UK). For removing the cells, the cells were washed with PBS and incubated for 2 minutes at 37°C with Trypsine (Gibco by Life technologies, Thermo Fischer, US).

Clonogenics assay

Cells were plated at very low densities (50/100/200 cells per well) in a 6-well plate and monitored for 3-5 days. The experiment was stopped before individual cell colonies started to merge and colonies were fixed and stained with 6% glutaraldehyde + 0,5% Crystal Violet solution (Sigma). Colonies were counted using a manual cell counter in ImageJ.

Protein extraction and Western Blotting

Protein was extracted by lysing the cells using a cold 1x radio immune-precitipation assay (RIPA) buffer that was prepared from a 10x stock solution (Pierce Thermo scientific, Rockford, USA). The lysed cells are collected and vortexed every 5 minutes for 30 minutes and spun down (13.000rpm x 10min, 4°C). The supernatant (protein) is carefully taken off. The protein is kept at -80°C until use.

Protein concentration was measured using a standard bicinchoninic acid (BCA) kit (Pierce Thermo scientific, Rockford, USA) and the FLUOstar Omega Plate reader (BMG Labtech). Equal amounts of protein were loaded onto a 15% Tris polyacrylamide gel and subjected to SDS polyacrylamide gel electrophoresis (BioRad) for 30 min on 70V and 115 min (approx..) on 110V at RT. Transfer of separated proteins on to nitrocellulose membrane was performed at 70V for 55 minutes at 4°C (ice block in tank). After blocking with 5% (little less, 2gr in 50ml) milk, membranes were probed for LC3 levels using a rabbit anti-human LC3 polyclonal antibody (1:500, Abgent, San Diego, US) followed by goat anti-rabbit IgG conjugated to horseradish peroxidase (HRP) and ECL (Millipore corporation, Billerica, USA) and performed chemiluminescence detection with the G:Box system (SynGene, Cambridge, UK).

Immunocytochemistry

Cells were grown and treated with different concentrations of Torin-1 on collagen I coated glass slides (boroscillicate glass, 13mm, VWR, thickness nr. 1) in a 24-well plate. Cells were fixed in 4%

paraformaldehyde for 20 minutes at RT / in 100% Methanol for 20 minutes at -20°C. After blocking and permeabilizing with 5% goat serum and 0,1% Triton-X-100 for 1 hour on RT, cells were incubated with a primary antibody Protein Light Chain 3 (LC3, 1:100, Abgent APG8B, San Diego, CA) overnight at 4°C. After washing with PBS, cells were incubated with appropriate secondary antibodies for 1 hour at RT. After washing with PBS, the glass slides were mounted on glass coverslips (26x76x1.0mm, Surgipath Leica Biosystems, Peterborough, UK) using mounting medium with DAPI (Vectashield, Vector Laboratories, Burlingame, CA) and observed under a Leica DMi8 inverted microscope (Leica, Wetzlar, Germany).

Senescence-associated β-Galactosidase assay

Senescence-associated β -Galactosidase activity is a well-documented feature of senescent cells (Itahana et al., 2007). The Senescence β-Galactosidase Staining Kit (Cell Signalling Technology, MA, USA) was used to detect β-galactosidase activity at pH 6, according to the manufacturer’s

instructions. ECFCs were cultured in collagen coated 24-well plates with an initial 3 x 104 cells per

(8)

well, and then left for 4-12 hours before treatment. To cause senescence, the cells were treated for at least 3 days with 0.2 µM Etoposide. The cells were washed with PBS before fixative solution (300 µL/well) was added and incubated at RT for 10-15 minutes to fix. The cells were washed two more times with PBS (gently) and then incubated with X-gal staining solution (20 mg/mL, Cell Signalling Technology, MA, USA) at 37 ºC overnight. After incubation, the X-gal solution was replaced by PBS and images were taken the next day with a Leica DMi1 and quantified manually using ImageJ.

Senescence was quantified by counting the amount of β-gal-positive cells and dividing that by the total amount of cells. Data are mean % of senescence ±SEM.

Flow Cytometry

Cells were plated on collagen coated T75 flasks and treatment started 4 hours after, when cells were attached. Cells were treated with either EBM-2 (only Primocin of all supplements) 2,5%FBS

(“starved”), EBM-2 2,5%FBS and 50ng/ml VEGF (“VEGF”), EGM-2 with all growth factors (GFs) and 10%FBS (“Control”), EGM-2 with all GFs and 20%FBS (“overfed”) for 3 days. Each condition had two flasks, one to stained and one unstained control. After treatment, the cells were stained with the Cell Proliferation Dye eFluor 450 (eBioscience, Altrincham, UK) following manufacturer’s protocol. After staining, cells were resuspended in 500 μl FACS buffer for analysis using a flow cytometer (Attune NxT, Life Technologies, Thermo Fischer, US) with analysis software (FlowJo, LLC; Version 10).

Respective unstained controls were used to determine accurate settings for data analysis.

Senescence conversion by Torin-1 experiment

For the experiments in which the effect of Torin-1 is tested on senescent ECFCs, the two agents were given in different combinations to the cells. In the β-gal Etoposide/torin-1 experiment the cells were given either no, or 30nM or 100nM Torin-1 pre-treatment for 24 hours before given a 3-day

treatment of 0.2microM Beta-Gal, or given no pre-treatment and the combination of 30nM Torin-1 and 0.2microM Etoposide for 3 days. Senescence was measured with β-gal (see ‘Senescence- associated β-Galactosidase assay’). In the clonogenics Etoposide/Torin-1 experiment, ECFCs were treated the same, only the Etoposide was given for 7 days and one more group was added which got Torin-1 áfter Etoposide treatment. Colonies were counted as described in the paragraph ‘Clonogenics assay’.

Cell Counting

For counting cells, the CASY Cell counter (Innovatis, Roche, Sussex, UK) was used. Cell suspension from a T25/T75 is dissolved in 1ml/3ml medium respectively. 50 μl is taken out and measured by the CASY Cell counter, dissolved in CASY cell counting buffer, following instructions. The machine then gives several values; viable cells/ml, total cells/ml and the mean diameter of the cells are the ones used for this experiment. The CASY provides a reading, which is the mean of three technical replicates of over thousands of cells.

Cell Metabolism assay (Seahorse)

To measure the effect of Torin-1 on ECFC metabolism, the Seahorse XFe96/24 Analyzer is used.

20.000 ECFCs/well were grown in 96-well plates (Seahorse Bioscience, Billerica, MA) in a 37C incubator in Seahorse basal medium (Seahorse Bioscience, Billerica, MA) supplemented with 10%

FBS. Two tests were performed: the mitochondrial stress (Mitostress) test and the effect on the oxygen consumption rate (OCR). For the Mito stress test, cells were pre-treated with different concentrations (30, 100, 300nM) Torin-1 for 12 hrs. After treatment, the cells were washed and incubated in a CO2-free chamber for 30 minutes before loaded into the Seahorse machine. The Mito stress was measured by serial injections of the Mito Stress Kit to the pre-treated cells: oligomycin (3 μM), FCCP (carbonyl cyanide 4-trifluoromethoxy-phenylhydrazone, 0,5 μM), rotenone (0.5 µM) plus antimycin A (0.5 µM) and vehicle. The injection cycles consisted of 3 minutes mixing time and 3 minutes measuring time after each injection.

(9)

For measuring the acute effects of Torin-1 on the OCR, no pre-treatment is given to the cells, but three consecutive injections of different Torin-1 concentrations (3x30nM, 3x100nM, 3x300nM or an increasing amount 30nM, 100nM, 300nM) followed by FCCP (4th injection, 0. 5 μM) were injected into the wells and measured trough the same mixing/measurement cycles. Data was normalized to the last measurement before the first injection (% OCR of baseline).

Reagents

Torin-1 was purchased from Selleckchem (Houston, US). Etoposide was purchased from Sigma- Aldrich (Saint Louis, US). Stock solutions were prepared in DMSO.

Statistical Analysis

All data are expressed as mean ±SEM. Statistical significance was evaluated by one-way ANOVA (and two-way ANOVA for the Seahorse experiment) with Bonferroni’s Post-hoc test (Prism version 5.0 for Mac; GraphPad Software, San Diego, US).

(10)

Results

Torin-1 has a cytostatic effect on proliferating ECFCs

To see what the dual mTOR inhibitor Torin-1 does to proliferating ECFCs, we tested the effect of Torin-1 in different concentrations on early passage ECFCs. To measure cytostatic effects, proliferating ECFCs were treated with different concentrations of Torin-1 (Fig. 5). After a 5-day treatment, drugs were washed out and colonies were stained with Crystal Violet. Torin-1 treatment significantly decreased size and number of ECFC colonies at all concentrations tested (>75%

decrease, p < 0.001). Torin-1 decreased ECFCs clonogenicity at the dose range 0.1µM to 10µM. Since all doses showed similar effect, we performed further experiments using lower Torin-1 doses (10- 100nM). Interestingly, this experiment using the automated cell counter CASY demonstrated a dose response (Fig. 6).

When the cells were incubated with lower concentrations of Torin-1 for 3 days and measured with the CASY cell counter, evidence shown demonstrated a decrease in cell number in response to an increasing dose of Torin-1 (Fig. 6).

- - - -

-

Control 0.1 μM 1 μM 10 μM

A

Figure 5. Response in colony formation to a 5-day treatment of different doses of Torin-1 (A) Representative image of

clonogenic assays (B)

Quantification of colonies. Data are mean number of colonies per well

±SEM; n = 6 for all groups; ***, p <

0.001, ANOVA. Post hoc comparisons with a Bonferroni test to compare means of each group with the control.

B

Figure 6. Cell number response to Torin-1.

Number of total cells in response to a 3-day treatment of different concentrations of torin-1, as measured by the CASY cell counter. Each value is made up out of 3 independent measurements of over thousands of cells (technical replicates), giving a relatively accurate indication of the effect of Torin-1 on ECFCs.

Cell number response to Torin-1

0 500000 1000000 1500000 2000000 2500000

# cells

Clonogenics response to torin-1

Control 0.1µ

M torin

M Torin 10µ

M torin 0

10 20 30

***

Dose

# colonies

***

*** ***

(11)

As an alternative approach to investigate effects of Torin-1 on proliferation, flow cytometry was used. Experimental principle is based on a fluorescent dye that halves in intensity each time a cell divides. Figure 7 confirmed results from figure 5 and 6, as the difference in positively stained cells and the median difference between stained and unstained cells becomes bigger with increasing concentrations of Torin-1.

All together, these data demonstrate that Torin-1 has a negative effect on ECFC proliferation. This indicates that Torin-1, when added to ECFCs in low concentrations, decreases cell proliferation.

Control (unstained vs. stained)

10nM torin treated cells

30nM torin treated cells

100nM torin treated cells

Figure 7. Flow cytometry results showing the effect of different concentrations of Torin-1 on proliferation of ECFCs, measured with eFluor 450 proliferation Dye which halved in intensity every time a cell divides and therefore is higher when proliferation is decreased.

Red peak = unstained control, blue peak = stained.

In the corner the percentage of positive stained cells compared to the unstained control and also the median difference shown.

(12)

Torin-1 and ECFCs senescence

To investigate effects of the mTOR inhibitor on senescence, we measured the average cell diameter of the ECFCs after a 3-day treatment of different concentrations of Torin-1. Figure 8 indicates that Torin-1 increases the mean diameter of ECFCs. Increase in cellular size is a characteristic of ECFCs that undergo senescence.

Following the β-Gal staining, a well-established marker to identify senescent cells, the effect of Torin- 1 on senescence is variable (Fig. 9). The control group exhibited ~50% of positively stained cells. A higher concentration of Torin-1 (≥1µM) seems to induce an increase in β-gal positive cells (65% and 60%). Data suggests that 1µM-10µM Torin-1 increases β-Gal staining in ECFCs.

Figure 8. Mean cell diameter of ECFCs in response to Torin-1. After a 3-day treatment of different dosages of Torin-1, the CASY cell counter measures the average ECFC diameter.

n (control) = 2137000, n (10nM) = 1438000, n (30nM) = 741800, n (100nM) = 520000.

Figure 9. Senescence response of ECFCs to Torin-1, after a 3-day treatment of different concentrations.

When the cells become senescent, β-gal-positive staining occurs. β-gal staining is quantified by dividing the amount of β-gal-positive cells by the total amount of cells. n = 6 for all groups. Post hoc comparisons with a Bonferroni test to compare means of each group. *, p < 0.05; **, p < 0,01, ANOVA.

Cell diameter in response to torin-1

Control 10nM 30nM 100nM

14 16 18 20 22

condition

mean diameter (µm)

Senescence in response to Torin-1

Control 0.1µ

M torin

M torin

10µM torin 0

20 40 60 80 100

Treatment

% senescent cells

*

**

(13)

Torin-1 effect on stress-induced senescent ECFCs

To examine the effect of Torin-1 on stress-induced senescence, we used the DNA damaging agent Etoposide. Different combinations of Etoposide and Torin-1 were tested on ECFCs: one group received Etoposide (0.2μM) only, two groups received two different concentrations (30/100nM) of Torin-1 as a pre-treatment for 24hrs before receiving Etoposide; and one group received 30nM Torin- 1 during Etoposide treatment (‘Torin+Etop’). When Torin-1 is added to ECFCs as a pre-treatment, there is a lower percentage β-gal positive senescent cells, with the 100nM Torin-1 treatment being significant (±60% decrease compared to Etop, p<0.001, Fig. 10). When Torin-1 is given

simultaneously with Etoposide, the inhibiting effect on senescence establishment is even stronger (±85% decrease compared to Etop, p<0,001). Interestingly, Torin-1 also significantly decreases the total number of cells when used together with Etoposide (also when comparing to the Etoposide group, Fig. 11). This suggests that Torin-1 might be inducing cell death in senescent ECFCs.

Control

Etoposide

30nM torin, Etop100nM torin, Etop

Torin + Etop 0

10 20 30

40

***

**

** ***

Etoposide / Torin-1 effects on senescence

Treatment

% senescent cells

Etoposide/Torin-1 effects on total cell number

Control

Etoposide Torin-1

30nM torin, Etop100nM torin, Etop Torin + Etop 0

100 200 300 400

500 #

*** ***

Treatment

# cells

Figure 10. Torin-1 lowers Etoposide-induced beta-gal staining. ECFCs were treated with 0.2 μM Etoposide for 4 days with groups given Torin- 1 as a pre-treatment or during Etoposide.

Quantified as the number of cells stained positive for beta-gal divided by the total number of cells. n

= 6 for all groups, except Etoposide group n (Etop)

= 18. Data are mean % senescent cells ±SEM; Post hoc comparisons with a Bonferroni test to compare means of each group. **, p<0.01, ***, p<0,001, ANOVA.

Figure 11. Torin-1 is not beneficial for the total cell number of Etoposide treated ECFCs, indicated by the low total cell numbers in the conditions in which Etoposide and Torin-1 were combined.

n = 6 for all groups, except Etoposide group n (Etop) = 18. Shown by the hash, the control group is sign. different from all groups (p<0.001). Data are mean number of cells ± SEM; Post hoc comparisons with a Bonferroni test to compare means of each group. ***, p<0,001, ANOVA.

(14)

Figure 12 shows the results of a clonogenics assay which includes a new group where Torin-1 (30nM) was added for 24hrs áfter Etoposide treatment (“Etop, Torin”). The clonogenics assay showed a slightly positive effect of Torin-1 pre-treatment and Torin-1 given during Etoposide treatment on colony number, whereas the group in which Torin-1 was given after Etoposide shows a lower amount of colonies.

Torin-1 effectively activates autophagy molecular machinery in ECFCs

To show the molecular mechanism to explain Torin-1 effects on ECFC proliferation and senescence, we performed Western Blotting for the protein Light Chain 3 (LC3), a well-established marker of autophagy. Tracking the conversion of LC3-I to LC3-II is indicative of autophagic activity. Data shows a decrease in LC3-I coupled with an increase in LC3-II, suggesting that Torin-1 activates autophagy in ECFCs.

LC3 immunocytochemistry (ICC) was performed in ECFCs that received 3 different concentrations of Torin-1 for 24 hours (Fig. 14). When autophagy is activated, LC3 is less equally dispersed over the cell surface and appears clustered. As shown in Figure 14, punctate staining of LC3 was present in all groups. Although none of the data points are significantly different, the analysis of the LC3 ICC showed that when the Torin-1 concentration is higher, LC3 appears in more clustered punctates (Fig.

15). These clusters were measured based on two different sizes, 50-500 and 100-500 pixel units (px2), and both showed an increase when Torin-1 increased, pointing at a higher number of

autophagosomes.

Clonogenics assay

Control

Etoposide Torin, EtopTorin + Etop Etop, Torin 0

20 40 60 80 100

*

#

Treatment

# colonies

A. Control 30nM

Control 30nM LC3-I

LC3-II

Β-actin

B. Control 30nM 100nM

Control 30nM 100nM LC3-I

LC3-II

Β-actin

Figure 12. Clonogenics results in response to Etoposide and/or Torin-1. Effect of different treatments on the colony-forming capacity of the cells, data are numbers of the quantification of colonies counted after treatment. n=6 for each group. Shown by the hash, the control group is sign.

different from all groups (p<0.05). Post hoc comparisons with a Bonferroni test to compare means of each group. *, p < 0.05, ANOVA.

Fig. 13. Western Blots show torin-1 works through activating autophagy. (A) and (B) show approximately the same, only in (B) an extra concentration of Torin-1 is tested. Both Western blots show a clear decrease in LC3-I and also a slight increase in LC3-II, indicating that Torin-1 activates autophagy. Β-actin was used as a loading control.

(15)

LC3 immunocytochemistry

Control 30nM torin

100nM torin 1000nM torin

Figure 14. LC3 Immunocytochemistry shows increased autophagy in Torin-1 treated ECFCs. Representative images of LC3 ICC staining of ECFCs treated with different concentrations of Torin-1 DAPI (blue) was used to visualize the nuclei, punctate staining of LC3 (red) was present in all groups but more clustered in the groups treated with Torin-1, indicating the activation of autophagy in these groups.

Figure 15. LC3 immunocytochemistry shows an increasing autophagy response to a growing concentration of torin-1 to ECFCs in culture. LC3 staining shows an increase in larger LC3 fluorescent clusters, indicating autophagy. Two different cluster sizes are used for analysis. Left: counted LC3 dots of size 30-500 pixel units, right: counted LC3 dots of size 50-500 pixel units. Both graphs show no significant differences.

Autophagy response to Torin-1 (50-500 pixels)

Control

30nM Torin 100nM Torin 1000nM Torin 0.0

0.5 1.0 1.5 2.0 2.5

Condition

# LC3 punctates of 50-500 pixels / cell

Autophagy response to Torin-1 (30-500 pixels)

Control

30nM Torin 100nM Torin 1000nM Torin 0

1 2 3 4

Condition

# LC3 punctates of 30-500 pixels / cell

(16)

Torin-1 reduces oxygen consumption in ECFCs

To show the effect of Torin-1 on ECFC functionality, we assessed the Torin-1 treated cells using the Seahorse SF Analyser, to measure the oxygen consumption rate (OCR) of cells. The OCR is an indicator for mitochondrial respiration and provides a valuable insight into the metabolic state of cells. The graph shows that the OCR in all Torin-treated groups had OCRs below the vehicle group (Fig. 16). Interestingly, the group which received an increasing amount of Torin-1 (dark blue) consistently exhibited the lowest OCR. Especially in the measurements after the 3rd injection, the group exhibited the lowest OCR values with the biggest difference compared to the vehicle group (red)(Fig. 17). After adding the FCCP, a stimulator of the OCR, all Torin-1 treated groups have a significantly lower OCR compared to the vehicle group each timepoint (Fig. 18). This suggests that ECFCs OCR is responsive to Torin-1, especially when given in increasing amounts, and that Torin-1 induces a decrease on mitochondrial respiration.

Figure 16. Effect of different torin-1 doses on the mitochondrial respiration in ECFCs. The mitochondrial respiration is depicted as the Oxygen Consumption Rate (OCR), measured by the Seahorse XF Analyser. n=10 for each group, except n(incr.

torin, dark blue)=11. Time in minutes on the X-axis and the OCR is given in percentage compared to the baseline, being the measurement before the first injection.

Figure 17. The OCR of the group that received increasing Torin-1 differs the most from control in the 3 timepoints after the 3rd injection. Graph shows that the ‘increasing Torin’-group is significantly lower than the vehicle group in e

ach measurement

Oxygen consumption rate (OCR) in response to acute treatments of Torin-1

OCR in response to Torin-1 at timepoints after 3rd injection

61min 67min 74min

0 50 100

150 Vehicle

30nM Torin 100nM Torin 300nM Torin Increasing Torin

*** *** ***

Timepoint

OCR % of baseline

*** * *

(17)

The Mito Stress experiment, in which ECFCs pre-treated with different concentrations of Torin-1 were sequentially exposed to Oligomycin, FCCP, and rotenone + antimycin A, did not highlight any differences between the groups (suppl. Fig 1).

Figure 18. Torin-treated groups show significantly lower OCR than the untreated control in the timepoints after the FCCP injection. Graph shows that in each timepoint after the FCCP injection, the OCR of all Torin-treated groups is significantly lower than the vehicle group. n=10 for each group, except n(incr. torin)=11. OCR is given in percentage compared to baseline (the measurement before the first injection) ±SD. Post hoc comparisons with a Bonferroni test to compare means of each group with the vehicle group. *, p<0.05, ***, p<0,001, ANOVA.

OCR in response to Torin-1 at timepoints after FCCP injection

81min 87min 94min

0 50 100

150 Vehicle

30nM Torin 100nM Torin 300nM Torin Increasing Torin

Timepoint

OCR % of baseline

** * ***** ********* *********

*** ***

(18)

Discussion

In this study, the effect of Torin-1 on ECFCs was investigated. ECFCs, as the “bona fine” endothelial progenitor cells, are expected to repair damaged vessels by closely interacting with resident capillary endothelium and re-establishing a functional endothelial barrier. The future potential therapeutic use of ECFCs in ischaemia can be of great importance for the fight against diabetic retinopathy. For clinical use, it is important that ECFCs can be expanded efficiently in vitro without the cells becoming senescent and losing proliferative potential.

mTOR inhibiting drugs such as rapamycin are currently of great interest with respect to their ability to slow down aging and increase lifespan (Chen et al., 2009; Harrison et al., 2009b; Zhang et al., 2014). Recent study by Leontieva et al. elucidated that the ATP-competitive mTOR inhibitor Torin-1, which targets both mTORC1 and less well-studied mTORC2, prevents geroconversion in a more efficient way than rapamycin (Leontieva et al., 2015). Torin-1 is therefore proposed as a potential anti-senescence drug.

This is the first study investigating the role of mTOR inhibitor Torin-1 on ECFC biology. To start characterising the Torin-1 effects on ECFCs, we first performed experiments of Torin-1 on healthy proliferating cells. These simple experiments showed that Torin-1 inhibits cell proliferation and upregulates senescence in proliferating ECFCs. Remarkable was the high percentage (~50%) of positively stained cells in the control group in figure 9. When 0,1 μM Torin-1 is added, senescence goes down (~40%), but when adding a higher concentration there seemed to be an increase in β-gal positive cells (65% and 60%). Although the control group was not of a particular late passage (passage 13), the percentage positively stained cells does indicate that the control group had, for some reason, become senescent. If so, this would suggest that a low dose Torin-1 trends to be beneficial in reversing senescence.

To elucidate whether Torin-1 is able to reverse or inhibit ECFC senescence, ECFCs were made senescent with the chemical agent Etoposide. Various methods and combinations of Torin-1 and/or Etoposide treatments gave interesting insights. Figure 10 showed that Torin-1 was able to diminish the Etoposide-induced senescence: 100nM Torin-1 pre-treatment was significantly lower compared to the Etop-group and the Torin treatment during Etop almost fully eliminated the senescence caused by Etoposide. However, figure 11 showed that total cell number of ECFCs also significantly decreased when Torin-1 is added, especially in combination with Etoposide. This suggests that the Torin-1 treatment facilitates the cell death of senescent cells, causing the loss of senescent cells, explaining both the lower β-gal staining and the lower cell number. It seems that in senescent ECFCs, Torin-1 inhibits an amount of senescence probably by inducing apoptosis. This observation fits well into the idea that mTOR is the signalling center of the pathway relevant to a senescence or apoptosis decision. Interestingly, rapamycin is found to enhance apoptosis in human and mouse cells treated with cisplatin (another senescence-inducing agent), although under basal conditions rapamycin does not promote apoptosis (Iglesias-Bartolome et al., 2012; Shi et al., 1995). Further experiments are warranted to confirm apoptosis as the cause for a lower cell number of senescent ECFCs in Torin-1 treated groups.

To answer the question through which mechanism Torin-1 has effect on proliferation and

senescence, we investigated whether Torin-1 activates autophagy. Western blot and ICC of LC3 were performed on Torin-1 treated ECFCs to see whether autophagy was upregulated. Autophagy involves a dynamic autophagosome formation and degradation that can be estimated by monitoring the protein levels of LC3, a microtubule-associated protein known to exist on autophagosomes (Kabeya et al., 2000). LC3 has two forms in the cell: LC3-I in the cytosol and LC3-II, which is associated specifically with the autophagosome membrane. The Western showed an increase in LC3-II and a lowering of LC3-I, which also happens to LC3 under starvation, indicating activation of autophagy

(19)

(Mizushima et al., 2004). The analysis of the LC3 immunocytochemistry required discussion on the interpretation of the LC3 clusters. According to guidelines, the number of LC3 puncta per cell, no matter the size, is used as a measure of autophagy. It is recommended not to use puncta size as a measurement for autophagy (Klionsky et al., 2016). However, in most literature the control has many small LC3 puncta as well, dispersed equally over the cell surface. These puncta become bigger or more intense after starvation or treatment with chloroquine (Lichtenstein et al., 2011; Mizushima, 2004). In these studies “LC3 clusters” and the “accumulation of LC3” is what indicates autophagy, making a difference between the bigger puncta to the small dots present in the control (see fig. 14, control). We used the size as a threshold to distinguish between control and Torin-1 treated cells, with the threshold clearly defined in the methods. This slight modification in the assessment methodology was required to clarify the difference between the puncta in controls and the bigger and brighter puncta in the treated groups. Some studies use the co-localisation of other autophagy- related proteins such as WIPI-1 and Atg9 in conjunction with LC3 as a functional readout of

autophagy (Proikas-Cezanne et al., 2007; Young et al., 2006). The use of other autophagy-proteins would be a valuable addition to our immunocytochemistry experiment.

Together, the LC3-I to LC3-II conversion in the Western Blot together with the increasing trend of larger autophagosomes indicated that Torin-1 effectively activates autophagy molecular machinery in ECFCs. Autophagy, the process by which cargo such as long-lived proteins and organelles is isolated and delivered to the lysosomes, is generally accepted as a mechanism to promote cell survival (Kenific & Debnath, 2015). Autophagy and death are both downstream of mTOR, cross- regulating each other usually in an inhibitory manner (Mattiolo et al., 2015). However, our findings suggest that in proliferating ECFCs, the induction of autophagy facilitates the activation of apoptosis (Fig. 5–7). Interestingly, in proliferating ECFCs the inhibition of mTOR also increased the amount of senescent cells (Fig. 8,9), indicating that mTOR inhibition can facilitate both. Still, it is unclear why certain cells go into senescence and certain cells go into apoptosis and which factors influence this decision. Furthermore, we have shown that Torin-1 seemed to have a different effect on stress- induced senescent cells; Torin-1 still lowered the cell number but also diminished senescence. This indicates that inhibiting mTOR and inducing autophagy activity might be beneficial in clearing senescent cells.

The last experiment of this article focused on finding the effects of Torin-1 of the function of ECFCs.

The Seahorse analyser interrogates mitochondrial respiration by measuring the oxygen consumption rate (OCR) of cells. We performed two experiments measuring the OCR; in the first experiment it was measured how the ECFCs responded to acute Torin-1 treatments of different concentrations, one group receiving an increasing concentration. As shown by figure 16, all the Torin-treated groups showed a lower OCR in comparison to the control. After the FCCP injection there was a significant difference between the vehicle and all Torin-treated groups (Fig. 18), indicating that Torin-1 inhibits mitochondrial respiration of ECFCs. This is in accordance with data of the effect of rapamycin on mitochondrial respiration, showing inhibition of mTOR by rapamycin decreased oxygen consumption and mitochondrial capacity (Cunningham et al., 2007; Ramanathan & Schreiber, 2009). mTORC1 has been demonstrated to control mitochondrial respiration(Morita et al., 2013). As Torin-1 inhibits both mTORC1 and 2, further experiments are needed in order to investigate whether this effect solely came from mTORC1 inhibition or mTORC2 also has influence on oxygen consumption.

Furthermore, the Increasing Torin group showed the lowest OCR, differing the most from the vehicle group. After the third injection, this difference is significant (Fig. 17). From this experiment it seems that injecting increasing Torin concentrations have more effect than adding constant low or high concentrations to ECFCs. A reason for this difference could be that it is more difficult for ECFCs to compensate and adjust to changing mTOR inhibition than to a consistent inhibition. However, the increasing Torin group already differed from the other groups after the first injection, in which the increasing Torin group received the same concentration (30nM) of Torin as the 30nM Torin group.

This suggests that the increasing Torin group had an intrinsic difference not caused by Torin-1.

(20)

Nonetheless, the Seahorse results indicate that Torin-1 has an effect of ECFC metabolism by inhibiting oxygen consumption. Following the widely believed theory that decreasing oxygen consumption decreases the production-rate of mitochondrial reactive oxygen species (ROS), Torin-1 would also have an effect on ROS. However, some studies have shown that mitochondrial oxygen consumption and ROS-production are modulated independently (Barja, 2007; Cortassa et al., 2014).

Apoptosis could also be induced in a mitochondria-dependent manner, which is modulated by i.a.

mitochondrial ROS formation (Pangare & Makino, 2012). To shed more light on these processes it would be of great interest to measure the effect of Torin-1 on ROS production.

Conclusion

In the search for treatments against diabetic retinopathy, ECFCs have come forward as the bona fide EPC: able to incorporate into vasculature and repair damaged vessels. For clinical use, these cells should be healthy and proliferating and mTOR inhibitor Torin-1 is a drug with the potential to slow aging down. Our results provide the first characterisation of Torin-1 effects on ECFCs. We established that adding Torin-1 to proliferating ECFCs induced senescence. However, the drug seems to have a gerosuppressive effect on stress-induced senescent ECFCs. Torin-1 has its effect through i.a.

autophagy activation, which has complex relations to senescence and apoptosis that should be further elucidated. Furthermore, mTOR inhibition with Torin-1 decreased mitochondrial respiration of ECFCs.

Further directions

Endothelial cells of diabetic patients are damaged due to the high glucose environment. Therefore a complementary approach to this experiment would be to see differences in ECFCs isolated from a diabetic patient and compare these to “healthy” ECFCs. Hyperglycemia is a primary contributor to the EPC number and it is found to induce premature senescence in EPCs (Chang et al., 2010; Fadini et al., 2007). This makes the ability of Torin-1 or any other mTOR inhibitor to reduce senescence of even bigger interest with regards to diabetic retinopathy. However, treatment with mTOR inhibitors has been shown to increase the chance on hyperglycemia and new-onset diabetes (Verges & Cariou, 2015). This makes mTOR inhibition unfavourable in diabetes on the one hand, but on the other hand it draws attention to further studying the function of mTOR in diabetes. Furthermore, would ECFCs from a patient with diabetes respond the same in terms of autophagy (LC3) and apoptosis? And would Torin-1 (or any similar mTOR inhibitor) have the ability to rescue the angiogenic potential of diabetic ECFCs?

mTOR is an important regulator of many processes involving lifespan regulation. There are many different mTOR inhibitors, all with a slightly different working mechanism, targeting different mTOR- complexes. Rapamycin is much better understood and has been used in a lot of research so far.

Therefore it would be a good idea to repeat these experiments for rapamycin, also to elucidate mTORC2 functions.

(21)

References

Ajlan, R. S., Silva, P. S., & Sun, J. K. (2016). Vascular Endothelial Growth Factor and Diabetic Retinal Disease.

Seminars in Ophthalmology, 31(1-2), 40-48.

Barja, G. (2007). Mitochondrial oxygen consumption and reactive oxygen species production are independently modulated: implications for aging studies. Rejuvenation Research, 10(2), 215-224.

Benjamin, D., Colombi, M., Moroni, C., & Hall, M. N. (2011). Rapamycin passes the torch: a new generation of mTOR inhibitors. Nature Reviews.Drug Discovery, 10(11), 868-880.

Boyer, D. S., Hopkins, J. J., Sorof, J., & Ehrlich, J. S. (2013). Anti-vascular endothelial growth factor therapy for diabetic macular edema. Therapeutic Advances in Endocrinology and Metabolism, 4(6), 151-169.

Chang, J., Li, Y., Huang, Y., Lam, K. S., Hoo, R. L., Wong, W. T., et al. (2010). Adiponectin prevents diabetic premature senescence of endothelial progenitor cells and promotes endothelial repair by suppressing the p38 MAP kinase/p16INK4A signaling pathway. Diabetes, 59(11), 2949-2959.

Chen, C., Liu, Y., Liu, Y., & Zheng, P. (2009). mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Science Signaling, 2(98), ra75.

Cortassa, S., O'Rourke, B., & Aon, M. A. (2014). Redox-optimized ROS balance and the relationship between mitochondrial respiration and ROS. Biochimica Et Biophysica Acta, 1837(2), 287-295.

Critser, P. J., & Yoder, M. C. (2010). Endothelial colony-forming cell role in neoangiogenesis and tissue repair.

Current Opinion in Organ Transplantation, 15(1), 68-72.

Cunningham, J. T., Rodgers, J. T., Arlow, D. H., Vazquez, F., Mootha, V. K., & Puigserver, P. (2007). mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. Nature, 450(7170), 736-740.

Dor, Y., Porat, R., & Keshet, E. (2001). Vascular endothelial growth factor and vascular adjustments to

perturbations in oxygen homeostasis. American Journal of Physiology.Cell Physiology, 280(6), C1367-74.

Dulic, V. (2013). Senescence regulation by mTOR. Methods in Molecular Biology (Clifton, N.J.), 965, 15-35.

Engerman, R. L. (1989). Pathogenesis of diabetic retinopathy. Diabetes, 38(10), 1203-1206.

Fadini, G. P., Sartore, S., Agostini, C., & Avogaro, A. (2007). Significance of endothelial progenitor cells in subjects with diabetes. Diabetes Care, 30(5), 1305-1313.

Harrison, D. E., Strong, R., Sharp, Z. D., Nelson, J. F., Astle, C. M., Flurkey, K., et al. (2009a). Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature, 460(7253), 392-395.

Harrison, D. E., Strong, R., Sharp, Z. D., Nelson, J. F., Astle, C. M., Flurkey, K., et al. (2009b). Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature, 460(7253), 392-395.

He, K., Lv, W., Zhang, Q., Wang, Y., Tao, L., & Liu, D. (2015). Gene set enrichment analysis of pathways and transcription factors associated with diabetic retinopathy using a microarray dataset. International Journal of Molecular Medicine, 36(1), 103-112.

Hirschi, K. K., Ingram, D. A., & Yoder, M. C. (2008). Assessing identity, phenotype, and fate of endothelial progenitor cells. Arteriosclerosis, Thrombosis, and Vascular Biology, 28(9), 1584-1595.

(22)

Hur, J., Yoon, C. H., Kim, H. S., Choi, J. H., Kang, H. J., Hwang, K. K., et al. (2004). Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology, 24(2), 288-293.

Iglesias-Bartolome, R., Patel, V., Cotrim, A., Leelahavanichkul, K., Molinolo, A. A., Mitchell, J. B., et al. (2012).

mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis.

Cell Stem Cell, 11(3), 401-414.

Itahana, K., Campisi, J., & Dimri, G. P. (2007). Methods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assay. Methods in Molecular Biology (Clifton, N.J.), 371, 21-31.

Kabeya, Y., Mizushima, N., Ueno, T., Yamamoto, A., Kirisako, T., Noda, T., et al. (2000). LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. The EMBO Journal, 19(21), 5720-5728.

Kang, S. A., Pacold, M. E., Cervantes, C. L., Lim, D., Lou, H. J., Ottina, K., et al. (2013). mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science (New York, N.Y.), 341(6144), 1236566.

Kenific, C. M., & Debnath, J. (2015). Cellular and metabolic functions for autophagy in cancer cells. Trends in Cell Biology, 25(1), 37-45.

Klionsky, D. J., Abdelmohsen, K., Abe, A., Abedin, M. J., Abeliovich, H., Acevedo Arozena, A., et al. (2016).

Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy, 12(1), 1-222.

Leontieva, O. V., & Blagosklonny, M. V. (2010). DNA damaging agents and p53 do not cause senescence in quiescent cells, while consecutive re-activation of mTOR is associated with conversion to senescence.

Aging, 2(12), 924-935.

Leontieva, O. V., Demidenko, Z. N., & Blagosklonny, M. V. (2015). Dual mTORC1/C2 inhibitors suppress cellular geroconversion (a senescence program). Oncotarget, 6(27), 23238-23248.

Lichtenstein, A., Minogue, P. J., Beyer, E. C., & Berthoud, V. M. (2011). Autophagy: a pathway that contributes to connexin degradation. Journal of Cell Science, 124(Pt 6), 910-920.

Lin, Y., Weisdorf, D. J., Solovey, A., & Hebbel, R. P. (2000). Origins of circulating endothelial cells and endothelial outgrowth from blood. The Journal of Clinical Investigation, 105(1), 71-77.

Luo, Y., Li, L., Zou, P., Wang, J., Shao, L., Zhou, D., et al. (2014). Rapamycin enhances long-term hematopoietic reconstitution of ex vivo expanded mouse hematopoietic stem cells by inhibiting senescence.

Transplantation, 97(1), 20-29.

Mattiolo, P., Yuste, V. J., Boix, J., & Ribas, J. (2015). Autophagy exacerbates caspase-dependent apoptotic cell death after short times of starvation. Biochemical Pharmacology, 98(4), 573-586.

Medina, R. J., O'Neill, C. L., Humphreys, M. W., Gardiner, T. A., & Stitt, A. W. (2010). Outgrowth endothelial cells: characterization and their potential for reversing ischemic retinopathy. Investigative Ophthalmology

& Visual Science, 51(11), 5906-5913.

Medina, R. J., O'Neill, C. L., O'Doherty, T. M., Chambers, S. E., Guduric-Fuchs, J., Neisen, J., et al. (2013). Ex vivo expansion of human outgrowth endothelial cells leads to IL-8-mediated replicative senescence and impaired vasoreparative function. Stem Cells (Dayton, Ohio), 31(8), 1657-1668.

Mizushima, N. (2004). Methods for monitoring autophagy. The International Journal of Biochemistry & Cell

(23)

Mizushima, N., Yamamoto, A., Matsui, M., Yoshimori, T., & Ohsumi, Y. (2004). In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker.

Molecular Biology of the Cell, 15(3), 1101-1111.

Mohamed, Q., Gillies, M. C., & Wong, T. Y. (2007). Management of diabetic retinopathy: a systematic review.

Jama, 298(8), 902-916.

Morita, M., Gravel, S. P., Chenard, V., Sikstrom, K., Zheng, L., Alain, T., et al. (2013). mTORC1 controls

mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation. Cell Metabolism, 18(5), 698-711.

Pangare, M., & Makino, A. (2012). Mitochondrial function in vascular endothelial cell in diabetes. Journal of Smooth Muscle Research = Nihon Heikatsukin Gakkai Kikanshi, 48(1), 1-26.

Proikas-Cezanne, T., Ruckerbauer, S., Stierhof, Y. D., Berg, C., & Nordheim, A. (2007). Human WIPI-1 puncta- formation: a novel assay to assess mammalian autophagy. FEBS Letters, 581(18), 3396-3404.

Ramanathan, A., & Schreiber, S. L. (2009). Direct control of mitochondrial function by mTOR. Proceedings of the National Academy of Sciences of the United States of America, 106(52), 22229-22232.

Reinisch, A., Hofmann, N. A., Obenauf, A. C., Kashofer, K., Rohde, E., Schallmoser, K., et al. (2009). Humanized large-scale expanded endothelial colony-forming cells function in vitro and in vivo. Blood, 113(26), 6716- 6725.

Roberts, A. C., & Porter, K. E. (2013). Cellular and molecular mechanisms of endothelial dysfunction in diabetes.

Diabetes & Vascular Disease Research, 10(6), 472-482.

Shi, Y., Frankel, A., Radvanyi, L. G., Penn, L. Z., Miller, R. G., & Mills, G. B. (1995). Rapamycin enhances apoptosis and increases sensitivity to cisplatin in vitro. Cancer Research, 55(9), 1982-1988.

Stewart, P. A., Hayakawa, K., Akers, M. A., & Vinters, H. V. (1992). A morphometric study of the blood-brain barrier in Alzheimer's disease. Laboratory Investigation; a Journal of Technical Methods and Pathology, 67(6), 734-742.

Urbich, C., & Dimmeler, S. (2004). Endothelial progenitor cells: characterization and role in vascular biology.

Circulation Research, 95(4), 343-353.

Verges, B., & Cariou, B. (2015). mTOR inhibitors and diabetes. Diabetes Research and Clinical Practice, 110(2), 101-108.

Williams, R., Airey, M., Baxter, H., Forrester, J., Kennedy-Martin, T., & Girach, A. (2004). Epidemiology of diabetic retinopathy and macular oedema: a systematic review. Eye (London, England), 18(10), 963-983.

Yoder, M. C., Mead, L. E., Prater, D., Krier, T. R., Mroueh, K. N., Li, F., et al. (2007). Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood, 109(5), 1801-1809.

Young, A. R., Chan, E. Y., Hu, X. W., Kochl, R., Crawshaw, S. G., High, S., et al. (2006). Starvation and ULK1- dependent cycling of mammalian Atg9 between the TGN and endosomes. Journal of Cell Science, 119(Pt 18), 3888-3900.

Yu, K., Toral-Barza, L., Shi, C., Zhang, W. G., Lucas, J., Shor, B., et al. (2009). Biochemical, cellular, and in vivo activity of novel ATP-competitive and selective inhibitors of the mammalian target of rapamycin. Cancer Research, 69(15), 6232-6240.

(24)

Zhang, Y., Bokov, A., Gelfond, J., Soto, V., Ikeno, Y., Hubbard, G., et al. (2014). Rapamycin extends life and health in C57BL/6 mice. The Journals of Gerontology.Series A, Biological Sciences and Medical Sciences, 69(2), 119-130.

(25)

Supplementary figures

Supplementary figure 1. Mitostress experiment on ECFCs pre-treated with different concentrations of torin-1 for 12 hrs show no clear difference between groups. Over time, the Oxygen Consumption Rate (OCR) is shown, with every few minutes another agent being added to the wells.

Referenties

GERELATEERDE DOCUMENTEN

Here we report on the experimental observation of weak localization in Le´vy glasses and compare our results with a recently developed theory for multiple scattering in

If London politicians are smart, then 2012 will be the year they start ensuring the benefits of London rule are as clear to the North East and Cumbria as the benefits of Union

showed acceptable loadings. Factor Analyses: Service Orientation and Execution. An exploratory factor analysis, using principal component analyses, was done on the 26

Sy sê dit is ’n groot uitdaging om die talent van jong professionele mense te bestuur wat baie meer ingelig is as wat die ouer garde op dieselfde ouderdom was.

A simple model of lateral momentum exchange between adjacent flow sections is used to calculate compartment-averaged flow velocities in a two-stage and in an

[8] defined eight phases of R&amp;D (from ‘target-to-hit’ to ‘submission to launch ’), and modeled costs per phase along the three most important independent cost drivers:

Figure 3.16 Structural homology model of CYP153A6 indicating the position of the mutants, corresponding to five groups based on protein expression and percentage activity

To what extent was AHSV maintained in the arid environment of the Khomas Region, through the distribution and abundance of its Culicoides vector and a possible cycling host,