Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Stem Cells ; 28(4): 810-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20201065

ABSTRACT

Mounting evidence indicates that mesenchymal stem cells (MSC) are pivotal to vascular repair and neointima formation in various forms of vascular disease. Yet, the mechanisms that allow MSC to resist apoptosis at sites where other cell types, such as endothelial cells (EC), are dying are not well defined. In the present work, we demonstrate that apoptotic EC actively release paracrine mediators which, in turn, inhibit apoptosis of MSC. Serum-free medium conditioned by apoptotic EC increases extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation and inhibits apoptosis (evaluated by Bcl-xL protein levels and poly (ADP-ribose) polymerase cleavage) of human MSC. A C-terminal fragment of perlecan (LG3) released by apoptotic EC is one of the mediators activating this antiapoptotic response in MSC. LG3 interacts with beta1-integrins, which triggers downstream ERK1/2 activation in MSC, albeit to a lesser degree than medium conditioned by apoptotic EC. Hence, other mediators released by apoptotic EC are probably required for induction of the full antiapoptotic phenotype in MSC. Adopting a comparative proteomic strategy, we identified epidermal growth factor (EGF) as a novel mediator of the paracrine component of the endothelial apoptotic program. LG3 and EGF cooperate in triggering beta1-integrin and EGF receptor-dependent antiapoptotic signals in MSC centering on ERK1/2 activation. The present work, providing novel insights into the mechanisms facilitating the survival of MSC in a hostile environment, identifies EGF and LG3 released by apoptotic EC as central antiapoptotic mediators involved in this paracrine response.


Subject(s)
Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Heparan Sulfate Proteoglycans/metabolism , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Apoptosis , Cells, Cultured , Endothelial Cells/cytology , Humans , MAP Kinase Signaling System , Mesenchymal Stem Cells/cytology
2.
J Biol Chem ; 283(40): 27220-9, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18658137

ABSTRACT

Proteolysis of extracellular matrix components and the production of cryptic bioactive factors play key roles in vascular remodeling. We showed previously that extracellular matrix proteolysis is triggered by the apoptosis of endothelial cells (EC), resulting in the release of an anti-apoptotic C-terminal fragment of endorepellin (LG3). Here, we characterize the endorepellin-cleaving proteases released by apoptotic EC using a multifaceted proteomics strategy. Cathepsin L (CathL), a cysteine protease known to be associated with cardiovascular disease progression in animal models and humans, was isolated from medium conditioned by apoptotic EC. CathL cleaved recombinant endorepellin in vitro, leading to LG3 release. Inhibition of CathL activity in EC exposed to pro-apoptotic stimuli prevented LG3 release without modulating the development of apoptosis in EC. Inhibition of caspase-3 activation in EC with the biochemical inhibitor DEVD-fluoromethyl ketone or small interfering RNAs concomitantly prevented CathL release by EC, LG3 production, and the development of paracrine anti-apoptotic activity. These data demonstrate that caspase-3 activation is a novel pathway of importance for triggering extracellular CathL release and the cleavage of extracellular matrix components.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans/metabolism , Peptide Fragments/metabolism , Animals , Cardiovascular Diseases/metabolism , Cathepsin L , Cell Line , Disease Models, Animal , Humans , Paracrine Communication/physiology , Peptides/metabolism , RNA, Small Interfering
3.
Nephrol Dial Transplant ; 22(2): 409-16, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17082211

ABSTRACT

BACKGROUND: Uraemia is associated with endothelial dysfunction, but the effect of uraemic plasma on the gene expression pattern of human coronary arterial endothelial cells (HCAEC) has never been defined. METHODS: HCAECs were exposed for 48 h to a culture medium supplemented with 20% uraemic vs normal plasma. We extracted mRNA and hybridized it onto Affymetrix HG-U133 Plus2 microarrays. We validated our findings for five genes of interest by real-time PCR and performed evaluations of cell proliferation and apoptosis in HCAECs exposed to uraemic vs normal plasma. RESULTS: Six genes involved in the regulation of cell-cycle progression (CDK-1, topoisomerase II, PDZ-binding kinase, CDCA1, protein SDP35, E2F transcription factor 8) and two genes of the cholesterol efflux system (ABCA1 and ABCG1) were down-regulated in HCAECs exposed to uraemic plasma (>1.75-fold change vs normal). Real-time PCR confirmed the down-regulation observed in the microarray experiment. Cell proliferation was significantly decreased in HCAECs exposed to uraemic vs normal plasma for 48 h (86 vs 95% of serum-starved control, P = 0.006). Exposure to uraemic plasma for 48 h was associated with increased apoptosis of HCAEC as compared with normal plasma (7.7 vs 2.8%, P < 0.001), a phenomenon that was further enhanced when oxidized LDLs (150 microg protein/ml) were added to the medium containing uraemic plasma (16.9 vs 7.7%, P < 0.001). CONCLUSIONS: The down-regulation of genes involved in cell-cycle progression and cholesterol efflux from HCAECs exposed to uraemic conditions could contribute to enhancing endothelial dysfunction and atherosclerosis in patients with chronic renal failure.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Coronary Vessels/pathology , Endothelium, Vascular/metabolism , Gene Expression/physiology , RNA/genetics , Uremia/blood , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/biosynthesis , Aged , Apoptosis/genetics , Cell Cycle/physiology , Cell Proliferation , Cells, Cultured , Coronary Vessels/metabolism , Female , Humans , Male , Microarray Analysis , Middle Aged , Plasma , Polymerase Chain Reaction , Uremia/pathology
4.
Biomaterials ; 28(6): 1209-17, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17129601

ABSTRACT

Complications following endovascular aneurysm repair (EVAR) are related to deficient healing around the stent-graft (SG). New generations of SG with surface properties that foster vascular repair could overcome this limitation. Our goal was to evaluate the potential of a new nitrogen-rich plasma-polymerised biomaterial, designated PPE:N, as an external coating for polyethylene terephtalate (PET)- or polytetrafluoro-ethylene (PTFE)-based SGs, to promote healing around the implant. Thin PPE:N coatings were deposited on PET and PTFE films. Then, adhesion, growth, migration and resistance to apoptosis of vascular smooth muscle cells (VSMCs) and fibroblasts, as well as myofibroblast differentiation, were assessed in vitro. In another experimental group, chondroitin sulphate (CS), a newly described mediator of vascular repair, was added to normal culture medium, to search for possible additional benefit. PPE:N-coatings, especially on PET, increased and accelerated cell adhesion and growth, compared with control PET and with standard polystyrene culture plates (PCP). PPE:N was also found to increase the resistance to apoptosis in VSMC, an important finding as aneurysms are characterised by VMSC depletion caused by a pro-apoptotic phenotype. Addition of CS in solution further increased migration and resistance to apoptosis. In conclusion, PPE:N-coating and/or CS could promote vascular repair around SGs following EVAR.


Subject(s)
Aneurysm/surgery , Blood Vessel Prosthesis/adverse effects , Endothelial Cells/drug effects , Nitrogen/administration & dosage , Stents/adverse effects , Wound Healing/drug effects , Aneurysm/complications , Aneurysm/pathology , Animals , Cell Line , Coated Materials, Biocompatible/administration & dosage , Coated Materials, Biocompatible/chemistry , Drug Implants , Endothelial Cells/pathology , Materials Testing , Nitrogen/chemistry , Rats
5.
J Biol Chem ; 281(41): 30383-92, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16882656

ABSTRACT

Dysregulation of apoptosis in endothelial cells (EC) and fibroblasts contributes to fibrosis. We have shown previously that apoptosis of EC triggers the proteolysis of extracellular matrix components and the release of a C-terminal fragment of perlecan, which in turn inhibits apoptosis of fibroblasts. Here we have defined the receptors and pathways implicated in this anti-apoptotic response in fibroblasts. Neutralizing alpha2beta1 integrin activity in fibroblasts exposed to either medium conditioned by apoptotic EC (SSC) or a recombinant perlecan C-terminal fragment (LG3) prevented resistance to apoptosis and is associated with decreased levels of Akt phosphorylation. Co-incubation of fibroblasts for 24 h with SSC or LG3 in the presence of PP2 (AG1879), a biochemical inhibitor of Src family kinases (SFKs) and focal adhesion kinase, showed a significantly decreased anti-apoptotic response. However, focal adhesion kinase gene silencing with RNA interference did not inhibit the anti-apoptotic response in fibroblasts. Src phosphorylation was increased in fibroblasts exposed to SSC, and transfection of fibroblasts with constitutively active Src mutants induced an anti-apoptotic response that was not further increased by SSC. Also, Src(-/-)Fyn(-/-) fibroblasts failed to mount an anti-apoptotic response in presence of SSC for 24 h but developed a complete anti-apoptotic response when exposed to SSC for 7 days. These results suggest that extracellular matrix fragments produced by apoptotic EC initiate a state of resistance to apoptosis in fibroblasts via an alpha2beta1 integrin/SFK (Src and Fyn)/phosphatidylinositol 3-kinase (PI3K)-dependent pathway. In the long term, additional SFK members are recruited for sustaining the anti-apoptotic response, which could play crucial roles in abnormal fibrogenic healing.


Subject(s)
Apoptosis , Fibrosis/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Heparan Sulfate Proteoglycans/chemistry , Integrin alpha2beta1/chemistry , src-Family Kinases/chemistry , Animals , Fibroblasts/metabolism , Humans , Mice , Mice, Transgenic , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , RNA Interference
6.
Biochem Biophys Res Commun ; 338(3): 1374-82, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16269129

ABSTRACT

Endothelial cells (EC) under stress release paracrine mediators that facilitate accumulation of vascular smooth muscle cells (VSCM) at sites of vascular injury. We found that medium conditioned by serum-starved EC increase proliferation and migration of VSCM in vitro. Fractionation of the conditioned medium followed by mass spectral analysis identified one bioactive component as vitamin D-binding protein (DBP). DBP induced both proliferation and migration of VSMC in vitro in association with increased phosphorylation of ERK 1/2. PD 98059, a biochemical inhibitor of ERK 1/2, abrogated these proliferative and migratory responses in VSMC. DBP is an important carrier for the vitamin-D sterols, 25-hydroxyvitamin-D, and 1alpha,25-dihydroxyvitamin-D. Both sterols inhibited the activity of DBP on VSMC, suggesting that vitamin D binding sites are important for initiating the activities of DBP on VSMC. Release of DBP at sites of endothelial injury represents a novel pathway favoring accumulation of VSMC at sites of vascular injury.


Subject(s)
Endothelial Cells/metabolism , Growth Substances/metabolism , Vitamin D-Binding Protein/metabolism , Amino Acid Sequence , Cell Movement , Cell Proliferation , Cells, Cultured , Culture Media, Serum-Free , Endothelial Cells/cytology , Growth Substances/chemistry , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Paracrine Communication , Phosphorylation , Solubility , Umbilical Cord/cytology , Umbilical Cord/metabolism , Vitamin D/pharmacology , Vitamin D-Binding Protein/chemistry
7.
J Immunol ; 174(9): 5740-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843576

ABSTRACT

Apoptosis of endothelial cells (EC) is appreciated as a primary pathogenic event in systemic sclerosis. Yet, how apoptosis of EC leads to fibrosis remains to be determined. We report that apoptosis of EC triggers the release of novel fibrogenic mediators. Medium conditioned by apoptotic EC (SSC) was found to inhibit apoptosis of fibroblasts, whereas medium conditioned by EC in which apoptosis was blocked (with either pan-caspase inhibition or Bcl-x(L) overexpression) did not. PI3K was activated in fibroblasts exposed to SSC. This was associated with downstream repression of Bim-EL and long-term up-regulation of Bcl-x(L) protein levels. RNA interference for Bim-EL in fibroblasts blocked apoptosis. SSC also induced PI3K-dependent myofibroblast differentiation with expression of alpha-smooth muscle actin, formation of stress fibers, and production of collagen I. A C-terminal fragment of the domain V of perlecan was identified as one of the fibrogenic mediators present in SSC. A synthetic peptide containing an EGF motif present on the perlecan fragment and chondroitin 4-sulfate, a glycosaminoglycan anchored on the domain V of perlecan, induced PI3K-dependent resistance to apoptosis in fibroblasts and myofibroblast differentiation. Human fibroblasts derived from sclerodermic skin lesions were more sensitive to the antiapoptotic activities of the synthetic peptide and chondroitin 4-sulfate than fibroblasts derived from normal controls. Hence, we propose that a chronic increase in endothelial apoptosis and/or increased sensitivity of fibroblasts to mediators produced by apoptotic EC could form the basis of a fibrotic response characterized by sustained induction of an antiapoptotic phenotype in fibroblasts and persistent myofibroblast differentiation.


Subject(s)
Apoptosis/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibroblasts/metabolism , Fibroblasts/pathology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Signal Transduction/immunology , Adult , Amino Acid Sequence , Cell Differentiation/immunology , Cell Line , Chondroitin Sulfates/pharmacology , Culture Media, Conditioned , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Fibroblasts/immunology , Heparan Sulfate Proteoglycans/physiology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Structure, Tertiary , Scleroderma, Systemic/immunology
8.
Am J Transplant ; 4(6): 946-52, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147429

ABSTRACT

A single-center cohort study of kidney and kidney-pancreas recipients was conducted to evaluate the association between new immunosuppressive regimens and risk of thrombotic microangiopathy (TMA). From January 1st,1996 to December 31, 2002, 368 patients received a kidney or kidney-pancreas transplant at our center. Four immunosuppressive regimens were evaluated as potential risk factors of TMA: cyclosporin + mycophenolate mofetil (CsA + MMF), cyclosporin + sirolimus (CsA + SRL), tacrolimus + myophenolate mofetil (FK + MMF), and tacrolimus + sirolimus (FK + SRL). Thirteen patients developed biopsy-proven TMA in the absence of vascular rejection. The incidence of TMA was significantly different in the four immunosuppressive regimens studied (p < 0.001). The incidence of TMA was highest in the CsA + SRL group (20.7%). The relative risk of TMA was 16.1 [95% confidence interval (CI): 4.3-60.8] for patients in the CsA + SRL group as compared with those in the FK + MMF group. We also investigated in vitro the pathophysiological basis of this association. The CsA-SRL combination was found to be the only regimen that concomitantly displayed pro-necrotic and anti-angiogenic activities on arterial endothelial cells. We propose that this combination concurs to development of TMA through dual activities on endothelial cell death and repair.


Subject(s)
Cyclosporine/adverse effects , Immunosuppressive Agents/adverse effects , Kidney Transplantation , Kidney/blood supply , Mycophenolic Acid/analogs & derivatives , Neovascularization, Pathologic/chemically induced , Pancreas Transplantation , Sirolimus/adverse effects , Thrombosis/chemically induced , Adult , Angiogenesis Inhibitors/pharmacology , Apoptosis , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/pathology , Cohort Studies , Drug Therapy, Combination , Endothelium, Vascular/drug effects , Graft Rejection/drug therapy , Humans , Middle Aged , Mycophenolic Acid/adverse effects , Necrosis , Risk Factors , Thrombosis/therapy
9.
FASEB J ; 18(6): 705-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14977881

ABSTRACT

Increased endothelial apoptosis and decreased apoptosis of vascular smooth muscle cells (VSMC) are central to initiation of myo-intimal thickening. We hypothesized that apoptosis of endothelial cells (EC) induces the release of anti-apoptotic mediator(s) active on VSMC. We found that serum-free medium conditioned by apoptotic EC decreases apoptosis of VSMC compared with fresh serum-free medium. Inhibition of endothelial apoptosis during conditioning with a pan-caspase inhibitor ZVAD-FMK blocked the release of the anti-apoptotic factor(s) active on VSMC. VSMC exposed to serum-free medium conditioned by apoptotic EC showed increased ERK 1/2 phosphorylation, enhanced Bcl-xl expression, and inhibition of p53 expression. Fractionation of the conditioned medium followed by mass spectral analysis identified one bioactive component as a C-terminal fragment of the domain V of perlecan. Serum-free medium supplemented with either a synthetic peptide containing the EGF motif of the domain V of perlecan or chondroitin 4-sulfate, a glycosaminoglycan anchored on the domain V of perlecan, increased ERK 1/2 phosphorylation and Bcl-xl protein levels while inhibiting apoptosis of VSMC. These results suggest that a proteolytic activity developing downstream of activated caspases in apoptotic EC initiates degradation of pericellular proteoglycans and liberation of bioactive fragments with a robust impact on inhibition of VSMC apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , Endothelium, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Paracrine Communication , Animals , Biological Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/physiology , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Tumor Suppressor Protein p53/metabolism , bcl-X Protein
10.
FASEB J ; 17(3): 515-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12514115

ABSTRACT

Blockade of the mitochondrial permeability transition pore (mPTP) by cyclosporin A (CsA) inhibits apoptosis in various cell types. However, use of CsA in humans is associated with damage to the arterial endothelium. We evaluated whether inhibition of the apoptotic machinery by CsA promotes other forms of cell death in arterial endothelial cells (EC). Exposure of human umbilical artery EC (HUAEC) to clinically relevant concentrations of CsA for up to 24 h was associated with a significant increase in necrotic features. We detected inhibition of apoptosis and a significant increase in necrosis in HUAEC exposed concomitantly to CsA and mitomycin C, a proapoptotic DNA damaging agent. We found that CsA-induced cell death is independent of caspase activation, p53 induction, and calcineurin inhibition. However, bongkrekic acid, another mPTP blocker, also increased necrosis in HUAEC. Dihydroethidium and acridine orange staining revealed increased intracellular production of reactive oxygen species (ROS) followed by lysosomal damage in HUAEC exposed to CsA. Hydroxyl radical and superoxide scavengers and inhibition of cathepsin D activity significantly attenuated CsA-induced EC death. These results suggest that inhibition of the apoptotic machinery by CsA in arterial EC favors development of a necrotic form of cell death regulated by ROS and secondary lysosomal damage.


Subject(s)
Apoptosis , Arteries/cytology , Cathepsin D/physiology , Cyclosporine/pharmacology , Endothelium, Vascular/cytology , Reactive Oxygen Species/metabolism , Caspases/metabolism , Cells, Cultured , DNA Damage , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Lysosomes/chemistry , Models, Biological , Necrosis , Oxidative Stress
11.
Transplantation ; 73(11): 1742-51, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12084996

ABSTRACT

BACKGROUND: Understanding the mechanisms of injury associated with cardiac arrest is essential for defining strategies aimed at improving preservation and function of kidneys harvested in non-heart-beating (NHB) donors. METHODS: We standardized a model of NHB donors in rats and studied the kinetics and types (apoptosis vs. necrosis) of renal cell death developing during cold storage. Using quantitative polymerase chain reaction, immunoblotting, and caspase inhibition, we also studied the molecular pathways regulating renal cell death in this model. RESULTS: The kinetics and extent of cell death developing in cortical tubules during cold storage were found to be increased in non-heart-beating (NHB) kidneys. Apoptosis of cortical tubules predominated in NHB kidneys exposed to 10 hr of cold storage, whereas necrosis increased after longer periods of cold ischemia. Shortly after cardiac arrest, a rapid up-regulation of Bax and Hsp 70 was found at the protein level in NHB kidneys. After 24 hr of cold storage, induction of Bax was maintained, whereas protein levels of Hsp70 returned to levels comparable to heart-beating (HB) controls. Also, mRNA levels of Bax were found to increase during cold storage in NHB kidneys. Cortical cell death was found to be largely caspase-independent but responsive to hydroxyl-radical scavenging with dimethyl sulfoxide (DMSO). CONCLUSIONS: Cardiac arrest promotes activation of death-inducing molecules such as Bax and is associated with increased development of caspase-independent renal cell death during cold storage. Developing strategies, such as free radical scavenging, aimed at inhibiting cell death during cold storage, could prove useful for improving preservation of NHB kidneys.


Subject(s)
Cell Death/physiology , Cold Temperature , Kidney Cortex/pathology , Kidney Transplantation , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , HSP70 Heat-Shock Proteins/genetics , Heart Arrest , Ischemia/pathology , Kidney Cortex/physiology , Male , Necrosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Inbred F344 , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein , bcl-X Protein
12.
Biochem Biophys Res Commun ; 291(2): 261-9, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11846399

ABSTRACT

The mechanisms of cytoprotection conferred by stress preconditioning remain largely uncharacterized in endothelial cells (EC). We report that stress preconditioning of EC with serum starvation induces the release of soluble mediator(s) that confer resistance to apoptosis, increase proliferation, and enhance angiogenesis in a second set of "non-preconditioned" EC. Preconditioning was found to target specifically the mitochondrial control of apoptosis in EC with increased protein levels of Bcl-2, decreased protein levels of Bax, and decreased cytosolic release of cytochrome c. Regulators of apoptosis acting upstream and downstream of the mitochondria such as p53, cIAP-1, cIAP-2, and XIAP were not altered. Mediators classically associated with preconditioning in other cell types such as adenosine, opioids, and nitric oxide are not implicated in this cytoprotective loop. Blockade of protein kinase C-dependent signaling inhibited cytoprotection of EC. Further characterization of this paracrine pathway should provide insights into the molecular regulation of preconditioning in endothelial cells.


Subject(s)
Apoptosis , Endothelium, Vascular/physiology , Paracrine Communication , Biological Factors/pharmacology , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free , Cytoprotection , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/physiology , Neovascularization, Physiologic , Protein Kinase C/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...