Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 49, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38365745

ABSTRACT

BACKGROUND: SMC1A is a subunit of the cohesin complex that participates in many DNA- and chromosome-related biological processes. Previous studies have established that SMC1A is involved in cancer development and in particular, is overexpressed in chromosomally unstable human colorectal cancer (CRC). This study aimed to investigate whether SMC1A could serve as a therapeutic target for CRC. METHODS: At first, we studied the effects of either SMC1A overexpression or knockdown in vitro. Next, the outcome of SMC1A knocking down (alone or in combination with bevacizumab, a monoclonal antibody against vascular endothelial growth factor) was analyzed in vivo. RESULTS: We found that SMC1A knockdown affects cell proliferation and reduces the ability to grow in anchorage-independent manner. Next, we demonstrated that the silencing of SMC1A and the combo treatment were effective in increasing overall survival in a xenograft mouse model. Functional analyses indicated that both treatments lead to atypical mitotic figures and gene expression dysregulation. Differentially expressed genes were implicated in several pathways including gene transcription regulation, cellular proliferation, and other transformation-associated processes. CONCLUSIONS: These results indicate that SMC1A silencing, in combination with bevacizumab, can represent a promising therapeutic strategy for human CRC.


Subject(s)
Cohesins , Colorectal Neoplasms , Animals , Humans , Mice , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Cell Cycle Proteins/metabolism , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , Cohesins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Silencing , Vascular Endothelial Growth Factor A/genetics
2.
Antioxid Redox Signal ; 27(6): 328-344, 2017 08 20.
Article in English | MEDLINE | ID: mdl-27960536

ABSTRACT

AIMS: Reactive oxygen species (ROS) play a pivotal role in different pathologic conditions, including ischemia, diabetes, and aging. We previously showed that ROS enhance miR-200c expression, causing endothelial cell (EC) apoptosis and senescence. Herein, we dissect the interaction among miR-200c and three strictly related proteins that modulate EC function and ROS production: sirtuin 1 (SIRT1), endothelial nitric oxide synthase (eNOS), and forkhead box O1 (FOXO1). Moreover, the role of miR-200c on ROS modulation was also investigated. RESULTS: We demonstrated that miR-200c directly targets SIRT1, eNOS, and FOXO1; via this mechanism, miR-200c decreased NO and increased the acetylation of SIRT1 targets, that is, FOXO1 and p53. FOXO1 acetylation inhibited its transcriptional activity on target genes, that is, SIRT1 and the ROS scavengers, catalase and manganese superoxide dismutase. In keeping, miR-200c increased ROS production and induced p66Shc protein phosphorylation in Ser-36; this mechanism upregulated ROS and inhibited FOXO1 transcription, reinforcing this molecular circuitry. These in vitro results were validated in three in vivo models of oxidative stress, that is, human skin fibroblasts from old donors, femoral arteries from old mice, and a murine model of hindlimb ischemia. In all cases, miR-200c was higher versus control and its targets, that is, SIRT1, eNOS, and FOXO1, were downmodulated. In the mouse hindlimb ischemia model, anti-miR-200c treatment rescued these targets and improved limb perfusion. Innovation and Conclusion: miR-200c disrupts SIRT1/FOXO1/eNOS regulatory loop. This event promotes ROS production and decreases NO, contributing to endothelial dysfunction under conditions of increased oxidative stress such as aging and ischemia. Antioxid. Redox Signal. 27, 328-344.


Subject(s)
Forkhead Box Protein O1/metabolism , MicroRNAs/genetics , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Sirtuin 1/genetics , Acetylation , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Oxidative Stress , Phosphorylation , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
3.
Oncotarget ; 7(47): 77257-77275, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27764787

ABSTRACT

Melanoma is the most aggressive skin-cancer, showing high mortality at advanced stages. Platelet Derived Growth Factor Receptor-alpha (PDGFR-alpha) potently inhibits melanoma- and endothelium-proliferation and its expression is significantly reduced in melanoma-biopsies, suggesting that melanoma progression eliminates cells expressing PDGFR-alpha. In the present study transient overexpression of PDGFR-alpha in endothelial (HUVEC) and melanoma (SKMel-28, A375, Preyer) human-cells shows strong anti-proliferative effects, with profound transcriptome and miRNome deregulation. PDGFR-alpha overexpression strongly affects expression of 82 genes in HUVEC (41 up-, 41 down-regulated), and 52 genes in SKMel-28 (43 up-, 9 down-regulated). CXCL10/IP-10 transcript showed up to 20 fold-increase, with similar changes detectable at the protein level. miRNA expression profiling in cells overexpressing PDGFR-alpha identified 14 miRNAs up- and 40 down-regulated, with miR-503 being the most down-regulated (6.4 fold-reduction). miR-503, miR-630 and miR-424 deregulation was confirmed by qRT-PCR. Interestingly, the most upregulated transcript (i.e., CXCL10/IP-10) was a validated miR-503 target and CXCL10/IP-10 neutralization significantly reverted the anti-proliferative action of PDGFR-alpha, and PDGFR-alpha inhibition by Dasatinb totally reverted the CXCL10/IP10 induction, further supporting a functional interplay of these factors. Finally, integration of transcriptomics and miRNomics data highlighted several pathways affected by PDGFR-alpha.This study demonstrates for the first time that PDGFR-alpha strongly inhibits endothelial and melanoma cells proliferation in a CXCL10/IP-10 dependent way, via miR-503 down-regulation.


Subject(s)
Chemokine CXCL10/genetics , Genomics/methods , MicroRNAs/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/cytology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Melanoma , Oligonucleotide Array Sequence Analysis
4.
J Biol Chem ; 287(53): 44761-71, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23148210

ABSTRACT

MicroRNA-210 (miR-210) induction is a virtually constant feature of the hypoxic response in both normal and transformed cells, regulating several key aspects of cardiovascular diseases and cancer. We found that miR-210 was induced in normoxic myoblasts upon myogenic differentiation both in vitro and in vivo. miR-210 transcription was activated in an hypoxia-inducible factor 1-α (Hif1a)-dependent manner, and chromatin immunoprecipitation experiments show that Hif1a bound to the miR-210 promoter only in differentiated myotubes. Accordingly, luciferase reporter assays demonstrated the functional relevance of the Hif1a binding site for miR-210 promoter activation in differentiating myoblasts. To investigate the functional relevance of increased miR-210 levels in differentiated myofibers, we blocked miR-210 with complementary locked nucleic acid oligonucleotides (anti-miR-210). We found that C2C12 myoblast cell line differentiation was largely unaffected by anti-miR-210. Likewise, miR-210 inhibition did not affect skeletal muscle regeneration following cardiotoxin damage. However, we found that miR-210 blockade greatly increased myotube sensitivity to oxidative stress and mitochondrial dysfunction. In conclusion, miR-210 is induced in normoxic myofibers, playing a cytoprotective role.


Subject(s)
Cell Differentiation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , Myoblasts/cytology , Myoblasts/metabolism , Oxygen/metabolism , Animals , Base Sequence , Cell Line , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Promoter Regions, Genetic
5.
Cell Transplant ; 21(9): 1997-2008, 2012.
Article in English | MEDLINE | ID: mdl-22469297

ABSTRACT

Presently, orthotopic liver transplant is the major therapeutic option for patients affected by primary liver diseases. This procedure is characterized by major invasive surgery, scarcity of donor organs, high costs, and lifelong immunosuppressive treatment. Transplant of hepatic precursor cells represents an attractive alternative. These cells could be used either for allogeneic transplantation or for autologous transplant after ex vivo genetic modification. We used stromal cells isolated from adipose tissue (AT-SCs) as platforms for autologous cell-mediated gene therapy. AT-SCs were transduced with lentiviral vectors expressing firefly luciferase, allowing for transplanted cell tracking by bioluminescent imaging (BLI). As a complementary approach, we followed circulating human α1-antitrypsin (hAAT) levels after infusion of AT-SCs overexpressing hAAT. Cells were transplanted into syngeneic mice after CCl(4)-induced hepatic injury. Luciferase bioluminescence signals and serum hAAT levels were measured at different time points after transplantation and demonstrate persistence of transplanted cells for up to 2 months after administration. These data, along with immunohistochemical analysis, suggest engraftment and repopulation of injured livers by transplanted AT-SCs. Moreover, by transcriptional targeting using cellular tissue-specific regulatory sequences, we confirmed that AT-SCs differentiate towards a hepatogenic-like phenotype in vitro and in vivo. Additionally, in transplanted cells reisolated from recipient animals' livers, we detected activation of the α-fetoprotein (AFP) promoter. This promoter is normally transcriptionally silenced in adult tissues but can be reactivated during liver regeneration, suggesting commitment towards hepatogenic-like differentiation of engrafted cells in vivo. Our data support AT-SC-mediated gene therapy as an innovative therapeutic option for disorders of liver metabolism.


Subject(s)
Adipose Tissue/cytology , Genetic Therapy/methods , Liver Diseases/surgery , Liver Regeneration/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Cell Differentiation , Humans , Liver Diseases/pathology , Mesenchymal Stem Cells/cytology , Mice , Tissue Distribution
6.
Arterioscler Thromb Vasc Biol ; 31(4): 898-907, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21233447

ABSTRACT

OBJECTIVE: Smad-interacting protein-1 (Sip1/ZEB2) is a transcriptional repressor of the telomerase reverse transcriptase catalytic subunit (Tert) and has recently been identified as a key regulator of embryonic cell fate with a phenotypic effect similar, in our opinion, to that reported for nitric oxide (NO). Remarkably, SIP1/ZEB2 is a known target of the microRNA 200 (miR-200) family. In this light, we postulated that Sip1/ZEB2 and the miR-200 family could play a role during the NO-dependent differentiation of mES. METHODS AND RESULTS: The results of the present study show that Sip1/ZEB2 expression is downregulated during the NO-dependent expression of mesendoderm and early cardiovascular precursor markers, including Flk1 and CXCR4 in mES. Coincidently, members of the miR-200 family, namely miR-429, -200a, -200b, and -200c, were transcriptionally induced in parallel to mouse Tert. This regulation occurred at the level of chromatin. Remarkably, miR-429/miR-200a overexpression or Sip1/ZEB2 knockdown by short hairpin RNA interference elicited a gene expression pattern similar to that of NO regardless of the presence of leukemia inhibitory factor. CONCLUSIONS: These results are the first demonstrating that the miR-200 family and Sip1/ZEB2 transcription factor are regulated by NO, indicating an unprecedented molecular circuitry important for telomerase regulation and early differentiation of mES.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Nitric Oxide/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Cell Differentiation/drug effects , Cells, Cultured , Chromatin Assembly and Disassembly , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Leukemia Inhibitory Factor/metabolism , Mice , Nitric Oxide Donors/pharmacology , RNA Interference , RNA, Messenger/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Telomerase/metabolism , Time Factors , Transcription, Genetic , Transfection , Zinc Finger E-box Binding Homeobox 2
7.
Cancer Res ; 70(23): 9711-20, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20952509

ABSTRACT

The CCAAT-binding transcription factor NF-Y plays a central role in regulating cellular proliferation by controlling the expression of genes required for cell-cycle progression such as cyclin A, cyclin B1, cyclin B2, cdc25A, cdc25C, and cdk1. Here we show that unrestricted NF-Y activity leads to apoptosis in an E2F1- and wild-type p53 (wtp53)-dependent manner. Unrestricted NF-Y activity induced an increase in E2F1 mRNA and protein levels. Furthermore, NF-Y directly bound the E2F1 promoter and this correlated with the appearance of open chromatin marks. The ability of NF-Y to induce apoptosis was impaired in cells lacking E2F1 and wtp53. Moreover, NF-Y overexpression elicited phosphorylation of wt p53Ser18 in an E2F1-dependent manner. Our findings establish that NF-Y acts upstream of E2F1 in p53-mediated apoptosis.


Subject(s)
Apoptosis/physiology , CCAAT-Binding Factor/physiology , E2F1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , E2F1 Transcription Factor/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , HCT116 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
8.
Aging (Albany NY) ; 2(8): 487-503, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20739737

ABSTRACT

Deregulated nutrient signaling plays pivotal roles in body ageing and in diabetic complications; biochemical cascades linking energy dysmetabolism to cell damage and loss are still incompletely clarified, and novel molecular paradigms and pharmacological targets critically needed. We provide evidence that in the retrovirus-packaging cell line HEK293-T Phoenix, massive cell death in serum-free medium is remarkably prevented or attenuated by either glucose or aminoacid withdrawal, and by the glycolysis inhibitor 2-deoxy-glucose. A similar protection was also elicited by interference with mitochondrial function, clearly suggesting involvement of energy metabolism in increased cell survival. Oxidative stress did not account for nutrient toxicity on serum-starved cells. Instead, nutrient restriction was associated with reduced activity of the mTOR/S6 Kinase cascade. Moreover, pharmacological and genetic manipulation of the mTOR pathway modulated in an opposite fashion signaling to S6K/S6 and cell viability in nutrient-repleted medium. Additionally, stimulation of the AMP-activated Protein Kinase concomitantly inhibited mTOR signaling and cell death, while neither event was affected by overexpression of the NAD+ dependent deacetylase Sirt-1, another cellular sensor of nutrient scarcity. Finally, blockade of the mTOR cascade reduced hyperglycemic damage also in a more pathophysiologically relevant model, i.e. in human umbilical vein endothelial cells (HUVEC) exposed to hyperglycemia. Taken together these findings point to a key role of the mTOR/S6K cascade in cell damage by excess nutrients and scarcity of growth-factors, a condition shared by diabetes and other ageing-related pathologies.


Subject(s)
Cell Survival/physiology , Food Deprivation/physiology , Intracellular Signaling Peptides and Proteins/physiology , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/physiology , Antimetabolites/administration & dosage , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free , Deoxyglucose/administration & dosage , HEK293 Cells , Humans , Mitochondria/physiology , Oxidative Stress/physiology , Ribosomal Protein S6/physiology , Ribosomal Protein S6 Kinases/physiology , Signal Transduction/physiology , Sirtuin 1/physiology , TOR Serine-Threonine Kinases/toxicity
9.
Stem Cells Int ; 2011: 304562, 2010 Dec 26.
Article in English | MEDLINE | ID: mdl-21234108

ABSTRACT

Chronic ulcers represent a major health problem in diabetic patients resulting in pain and discomfort. Conventional therapy does not guarantee adequate wound repair. In diabetes, impaired healing is partly due to poor endothelial progenitor cells mobilisation and homing, with altered levels of the chemokine stromal-derived factor-1 (SDF-1) at the wound site. Adipose tissue-associated stromal cells (AT-SCs) can provide an accessible source of progenitor cells secreting proangiogenic factors and differentiating into endothelial-like cells. We demonstrated that topical administration of AT-SCs genetically modified ex vivo to overexpress SDF-1, promotes wound healing into diabetic mice. In particular, by in vivo bioluminescent imaging analysis, we monitored biodistribution and survival after transplantation of luciferase-expressing cells. In conclusion, this study indicates the therapeutic potential of AT-SCs administration in wound healing, through cell differentiation, enhanced cellular recruitment at the wound site, and paracrine effects associated with local growth-factors production.

10.
PLoS One ; 4(8): e6603, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19668373

ABSTRACT

BACKGROUND: In the cardiovascular system, laminar shear stress (SS) is one of the most important source of endothelial protecting signals. Physical and chemical agents, however, including ionising radiations and anticancer drugs, may injure endothelial cells determining an increase in oxidative stress and genotoxic damage. Whether the SS protective function remains intact in the presence of strong oxidants or DNA damage is currently unclear. METHODS AND RESULTS: To investigate this aspect a series of experiments were performed in which HUVEC were exposed to sub-lethal doses of the radio-mimetic compound Bleomycin (Bleo; 10 microg/ml) which generated free radicals (ROS) without significantly compromising cell survival. Remarkably, the application of a SS of 12 dyne/cm(2) did not protect endothelial cells but markedly accelerated apoptosis compared to controls kept in static culture and in the presence of Bleo. Experiments with the inducible nitric oxide synthase (iNOS) inhibitor GW274150 significantly reduced the SS-dependent apoptosis indicating that the production of NO was relevant for this effect. At molecular level, the ataxia-telangectasia-mutated (ATM) kinase, the homeodomain-interacting protein kinase-2 (HIPK2) and p53 were found activated along a pro-apoptotic signalling pathway while p21(waf1,cip1,sdi1) was prevented from its protective action. RNA interference experiments revealed that HIPK2 and p53 were both important for this process, however, only the forced expression p21(waf1,cip1,sdi1) fully restored the SS-dependent pro-survival function. CONCLUSIONS: This study provides the first evidence that, in the presence of genotoxic damage, laminar flow contributes to endothelial toxicity and death and identifies molecular targets potentially relevant in endothelial dysfunction and cardiovascular disease pathogenesis.


Subject(s)
Carrier Proteins/metabolism , Endothelium, Vascular/enzymology , Protein Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Base Sequence , Bleomycin/pharmacology , Blotting, Western , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , DNA Primers , Endothelium, Vascular/cytology , Enzyme Activation , Humans , Polymerase Chain Reaction , RNA Interference , Reactive Oxygen Species , Tumor Suppressor Protein p53/metabolism
11.
FASEB J ; 23(7): 2131-41, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19264835

ABSTRACT

The present study provides evidence that abnormal patterns of global histone modification are present in the skeletal muscle nuclei of mdx mice and Duchenne muscular dystrophy (DMD) patients. A combination of specific histone H3 modifications, including Ser-10 phosphorylation, acetylation of Lys 9 and 14, and Lys 79 methylation, were found enriched in muscle biopsies from human patients affected by DMD and in late-term fetuses, early postnatal pups, or adult mdx mice. In this context, chromatin immunoprecipitation experiments showed an enrichment of these modifications at the loci of genes involved in proliferation or inflammation, suggesting a regulatory effect on gene expression. Remarkably, the reexpression of dystrophin induced by gentamicin treatment or the administration of nitric oxide (NO) donors reversed the abnormal pattern of H3 histone modifications. These findings suggest an unanticipated link between the dystrophin-activated NO signaling and the remodeling of chromatin. In this context, the regulation of class IIa histone deacetylases (HDACs) 4 and 5 was found altered as a consequence of the reduced NO-dependent protein phosphatase 2A activity, indicating that both NO and class IIa HDACs are important for satellite cell differentiation and gene expression in mdx mice. In conclusion, this work provides the first evidence of a role for NO as an epigenetic regulator in DMD.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Histones/metabolism , Muscular Dystrophy, Duchenne/pathology , Nitric Oxide/deficiency , Protein Processing, Post-Translational , Animals , Cell Nucleus , Humans , Mice , Mice, Inbred Strains , Muscle, Skeletal/pathology
12.
Proc Natl Acad Sci U S A ; 105(49): 19183-7, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19047631

ABSTRACT

The overlapping histological and biochemical features underlying the beneficial effect of deacetylase inhibitors and NO donors in dystrophic muscles suggest an unanticipated molecular link among dystrophin, NO signaling, and the histone deacetylases (HDACs). Higher global deacetylase activity and selective increased expression of the class I histone deacetylase HDAC2 were detected in muscles of dystrophin-deficient MDX mice. In vitro and in vivo siRNA-mediated down-regulation of HDAC2 in dystrophic muscles was sufficient to replicate the morphological and functional benefits observed with deacetylase inhibitors and NO donors. We found that restoration of NO signaling in vivo, by adenoviral-mediated expression of a constitutively active endothelial NOS mutant in MDX muscles, and in vitro, by exposing MDX-derived satellite cells to NO donors, resulted in HDAC2 blockade by cysteine S-nitrosylation. These data reveal a special contribution of HDAC2 in the pathogenesis of Duchenne muscular dystrophy and indicate that HDAC2 inhibition by NO-dependent S-nitrosylation is important for the therapeutic response to NO donors in MDX mice. They also define a common target for independent pharmacological interventions in the treatment of Duchenne muscular dystrophy.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Animals , Benzamides/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Histone Deacetylase 2 , Histone Deacetylases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Myoblasts/cytology , Myoblasts/enzymology , Nitric Oxide/metabolism , Nitrogen/metabolism , Pyridines/pharmacology , RNA, Small Interfering , Repressor Proteins/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/enzymology
13.
FASEB J ; 19(6): 629-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15705741

ABSTRACT

Laminar shear stress (LSS) represents a major athero-protective stimulus. However, the mechanisms for this effect are poorly characterized. As chemokine receptors modulate endothelial cell functions, we hypothesized that at least some LSS effects on endothelial cells (ECs) may be due to LSS-dependent changes in chemokine receptor expression and function. Exposure of Human umbilical vein endothelial cells (HUVECs) to 15 dynes/cm2/sec(-1) LSS strongly inhibited CXC chemokine receptor 4 (CXCR4) expression at the transcriptional level and impaired stromal-derived factor (SDF)-1/CXCL12-driven chemotaxis. On the contrary, low shear stress (SS; 4 dynes/cm2/sec(-1)) only marginally affected CXCR4 expression when compared with static control cells. Differently from CXCR4, the expression of SDF-1 mRNA was not affected by LSS treatment. CXCR4 overexpression induced a dose-dependent endothelial cell apoptosis that was enhanced by SDF-1 treatment and was caspase-dependent. CXCR4 overexpression inhibited the LSS-mediated antiapoptotic effect on ECs and was associated to impairment of LSS-induced ERK1/2 phosphorylation. These findings suggest that LSS-induced CXCR4 down-regulation may contribute to endothelial cell survival. Interestingly, the expression of the proatherogenic chemokines MCP-1 and IL-8 was induced by SDF-1 treatment and by CXCR4 overexpression in HUVECs. Further, the known LSS-induced inhibition of MCP-1 expression was impaired in CXCR4 overexpressing ECs. Finally, CXCR4 was abundantly expressed by human atherosclerotic plaque endothelium that is exposed to low/absent shear stress, while it was poorly expressed by minimally diseased carotid artery endothelium. In conclusion, LSS-dependent CXCR4 down-regulation may contribute to atheroprotection by favoring the integrity of the endothelial barrier and by inhibiting MCP-1 and IL-8 expression.


Subject(s)
Atherosclerosis/etiology , Endothelial Cells/physiology , Gene Expression , Hemorheology , Receptors, CXCR4/genetics , Receptors, CXCR4/physiology , Cell Survival , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CXCL12 , Chemokines, CXC/genetics , Chemokines, CXC/pharmacology , Chemokines, CXC/physiology , Chemotaxis , Endothelial Cells/chemistry , Gene Expression/drug effects , Humans , Interleukin-8/genetics , Microcirculation/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological , Umbilical Veins
14.
Cardiovasc Res ; 61(4): 693-704, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14985066

ABSTRACT

OBJECTIVE: The antiapoptotic effect of p21(Waf1/Cip1/Sdi1) (p21) was examined in human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress (SS) or to the nitric oxide donor sodium nitroprusside (SNP) and in a mouse model of hindlimb ischemia. METHODS: In vitro: Cells were cultured without serum and in the presence of cobalt chloride to simulate hypoxia for 12 h (T0). Shear stress was applied to endothelial cells for additional 12 h. In vivo: Hindlimb ischemia was realized in mice by femoral artery ligation. SNP was acutely administered by subcutaneous injection or by Alzet osmotic pumps for a longer treatment. RESULTS: At T0, HUVEC were either exposed to SS (15 dyn/cm2/s(-1)), treated with SNP or kept in static condition (ST) for 1-12 h; after additional 12 h in ST, 30-35% of cells still alive at T0 had died. In this condition, both SS and SNP treatments markedly increased p21 levels and reduced apoptosis in HUVEC. Recombinant adenoviruses carrying p21 (AdCMV.p21) or antisense p21 (AdCMV.ASp21) cDNA revealed that AdCMV.p21-infected HUVEC were protected from death while AdCMV.ASp21 reduced SS- and SNP-dependent protection from apoptosis. In mice, apoptosis was detected in endothelial cells of ischemic hindlimbs as early as 8 h after femoral artery ligation. Treatment with SNP enhanced p21 expression and protected ischemic tissue from damage. Remarkably, direct in vivo injection of AdCMV.p21 significantly reduced the number of apoptotic nuclei in the presence of ischemia. CONCLUSIONS: The present study establishes that, under our experimental conditions, (a) p21 plays an important role in SS and nitric oxide antiapoptotic effect in vitro, and (b) p21 gene transfer prevents apoptosis in vitro and in vivo, following acute interruption of blood flow.


Subject(s)
Cyclins/pharmacology , Endothelial Cells/drug effects , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA, Antisense/administration & dosage , Genetic Vectors/administration & dosage , Hindlimb , Humans , Ischemia/drug therapy , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Inbred Strains , Mice, Nude , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Stress, Mechanical , Transduction, Genetic
15.
Arterioscler Thromb Vasc Biol ; 22(3): 405-11, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11884282

ABSTRACT

In the present study, we analyzed the effect of conditioned media (CM) from bovine aortic endothelial cells exposed to laminar shear stress (SS) of 5 dyne/cm2 (SS5) or 15 dyne/cm2 (SS15) for 16 hours on smooth muscle cell (SMC) migration. In response to CM from bovine aortic endothelial cells exposed to SS5 (CMSS5) and SS15 (CMSS15), migration was 45 +/- 5.5 and 30 +/- 1.5 cells per field, respectively (P<0.05). Similar results were obtained with SS of 2 versus 20 dyne/cm2 and also when SS of 5 and 15 dyne/cm2 lasted 24 hours. Platelet-derived growth factor (PDGF)-AA levels in CMSS5 and CMSS15 were 9 +/- 7 and 18 +/- 5 ng/10(6) cells for 16 hours, respectively (P<0.05); PDGF-BB levels in CMSS5 and CMSS15 were 38 +/- 10 and 53 +/- 10 ng/10(6) cells for 16 hours, respectively (P<0.05). PDGF receptor alpha (PDGFRalpha) and PDGF receptor beta (PDGFRbeta) in SMCs were phosphorylated by CMSS15>CMSS5. In response to CMSS15, a neutralizing antibody against PDGF-AA enhanced SMC migration to a level comparable to that of CMSS5; in contrast, antibodies against PDGF-BB abolished SMC migration. Transfection of SMCs with a dominant-negative PDGFRalpha or PDGFRbeta increased or inhibited, respectively, SMC migration in response to CMSS15. Overexpression of wild-type PDGFRalpha inhibited SMC migration in response to CMSS5, CMSS15, or recombinant PDGF-BB (P<0.001). These results suggest that the ability of high SS to inhibit arterial wall thickening in vivo may be related to enhanced activation of PDGFRalpha in SMCs by PDGF isoforms secreted by the endothelium.


Subject(s)
Chemotaxis , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Platelet-Derived Growth Factor/biosynthesis , Animals , Becaplermin , Cattle , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/drug effects , Mutation , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/physiology , Stress, Mechanical , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...