Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 569: 216306, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37442366

ABSTRACT

Bidirectional interactions between cancer cells and their microenvironment govern tumor progression. Among the stromal cells in this microenvironment, adipocytes have been reported to upregulate cancer cell migration and invasion by producing fatty acids. Conversely, cancer cells alter adipocyte phenotype notably via increased lipolysis. We aimed to identify the mechanisms through which cancer cells trigger adipocyte lipolysis and evaluate the functional consequences on cancer progression. Here, we show that cancer cell-induced acidification of the extracellular medium strongly promotes preadipocyte lipolysis through a mechanism that does not involve lipophagy but requires adipose triglyceride lipase (ATGL) activity. This increased lipolysis is triggered mainly by attenuation of the G0/G1 switch gene 2 (G0S2)-induced inhibition of ATGL. G0S2-mediated regulation in preadipocytes affects their communication with breast cancer cells, modifying the phenotype of the cancer cells and increasing their resistance to chemotherapeutic agents in vitro. Furthermore, we demonstrate that the adipocyte-specific overexpression of G0S2 impairs mammary tumor growth and lung metastasis formation in vivo. Our results highlight the importance of acidosis in cancer cell-adipocyte crosstalk and identify G0S2 as the main regulator of cancer-induced lipolysis, regulating tumor establishment and spreading.


Subject(s)
Cell Cycle Proteins , Neoplasms , Cell Cycle Proteins/metabolism , Lipase/genetics , Lipase/metabolism , Adipocytes/metabolism , Lipolysis , Cell Physiological Phenomena
2.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36835422

ABSTRACT

RhoGDI2 is a guanine nucleotide dissociation inhibitor (GDI) specific for the Rho family of small GTPases. It is highly expressed in hematopoietic cells but is also present in a large array of other cell types. RhoGDI2 has been implicated in multiple human cancers and immunity regulation, where it can display a dual role. Despite its involvement in various biological processes, we still do not have a clear understanding of its mechanistic functions. This review sheds a light on the dual opposite role of RhoGDI2 in cancer, highlights its underappreciated role in immunity and proposes ways to explain its intricate regulatory functions.


Subject(s)
Immunity , Neoplasms , rho Guanine Nucleotide Dissociation Inhibitor beta , Humans , Neoplasms/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
3.
JCI Insight ; 7(8)2022 04 22.
Article in English | MEDLINE | ID: mdl-35316211

ABSTRACT

The capacity of ADAMTS3 to cleave pro-VEGFC into active VEGFC able to bind its receptors and to stimulate lymphangiogenesis has been clearly established during embryonic life. However, this function of ADAMTS3 is unlikely to persist in adulthood because of its restricted expression pattern after birth. Because ADAMTS2 and ADAMTS14 are closely related to ADAMTS3 and are mainly expressed in connective tissues where the lymphatic network extends, we hypothesized that they could substitute for ADAMTS3 during adulthood in mammals allowing proteolytic activation of pro-VEGFC. Here, we demonstrated that ADAMTS2 and ADAMTS14 are able to process pro-VEGFC into active VEGFC as efficiently as ADAMTS3. In vivo, adult mice lacking Adamts2 developed skin lymphedema due to a reduction of the density and diameter of lymphatic vessels, leading to a decrease of lymphatic functionality, while genetic ablation of Adamts14 had no impact. In a model of thermal cauterization of cornea, lymphangiogenesis was significantly reduced in Adamts2- and Adamts14-KO mice and further repressed in Adamts2/Adamts14 double-KO mice. In summary, we have demonstrated that ADAMTS2 and ADAMTS14 are as efficient as ADAMTS3 in activation of pro-VEGFC and are involved in the homeostasis of the lymphatic vasculature in adulthood, both in physiological and pathological processes.


Subject(s)
Lymphatic Vessels , Lymphedema , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , Animals , Homeostasis , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Lymphedema/genetics , Lymphedema/metabolism , Mammals/metabolism , Mice
4.
Sci Rep ; 8(1): 7050, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29728578

ABSTRACT

Propranolol, a widely used non-selective beta-adrenergic receptor blocker, was recently shown to display anticancer properties. Its potential to synergize with certain drugs has been also outlined. However, it is necessary to take into account all the properties of propranolol to select a drug that could be efficiently combined with. Propranolol was reported to block the late phase of autophagy. Hence, we hypothesized that in condition enhancing autophagy flux, cancer cells should be especially sensitive to propranolol. 2DG, a glycolysis inhibitor, is an anti-tumor agent having limited effect in monotherapy notably due to induction of pro-survival autophagy. Here, we report that treatment of cancer cells with propranolol in combination with the glycolysis inhibitor 2DG induced a massive accumulation of autophagosome due to autophagy blockade. The propranolol +2DG treatment efficiently prevents prostate cancer cell proliferation, induces cell apoptosis, alters mitochondrial morphology, inhibits mitochondrial bioenergetics and aggravates ER stress in vitro and also suppresses tumor growth in vivo. Our study underlines for the first time the interest to take advantage of the ability of propranolol to inhibit autophagy to design new anti-cancer therapies.


Subject(s)
Carbohydrate Metabolism/drug effects , Glucose/metabolism , Propranolol/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Autophagy/drug effects , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum Stress/drug effects , Glycolysis/drug effects , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays
5.
Oncotarget ; 6(13): 11264-80, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25834103

ABSTRACT

Lipogenesis inhibition was reported to induce apoptosis and repress proliferation of cancer cells while barely affecting normal cells. Lipins exhibit dual function as enzymes catalyzing the dephosphorylation of phosphatidic acid to diacylglycerol and as co-transcriptional regulators. Thus, they are able to regulate lipid homeostasis at several nodal points. Here, we show that lipin-1 is up-regulated in several cancer cell lines and overexpressed in 50 % of high grade prostate cancers. The proliferation of prostate and breast cancer cells, but not of non-tumorigenic cells, was repressed upon lipin-1 knock-down. Lipin-1 depletion also decreased cancer cell migration through RhoA activation. Lipin-1 silencing did not significantly affect global lipid synthesis but enhanced the cellular concentration of phosphatidic acid. In parallel, autophagy was induced while AKT and ribosomal protein S6 phosphorylation were repressed. We also observed a compensatory regulation between lipin-1 and lipin-2 and demonstrated that their co-silencing aggravates the phenotype induced by lipin-1 silencing alone. Most interestingly, lipin-1 depletion or lipins inhibition with propranolol sensitized cancer cells to rapamycin. These data indicate that lipin-1 controls main cellular processes involved in cancer progression and that its targeting, alone or in combination with other treatments, could open new avenues in anticancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Lipogenesis , Phosphatidate Phosphatase/metabolism , Prostatic Neoplasms/drug therapy , Sirolimus/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Male , Molecular Targeted Therapy , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/antagonists & inhibitors , Phosphatidate Phosphatase/genetics , Phosphorylation , Propranolol/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Ribosomal Protein S6/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
6.
Commun Integr Biol ; 5(1): 99-101, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22482023

ABSTRACT

The small GTPases of the Rho family are key signaling molecules regulating a plethora of biological pathways. They can exert diverse, sometimes opposite, contributions to specific cellular processes explaining why their regulation and their crosstalk must be finely tuned. Several mechanisms driving crosstalk between Rho GTPases have been described in the literature. They implicate proteins regulating their activity or common downstream effectors. Among the proteins regulating Rho GTPases cycling, RhoGDIs were viewed until very recently as passive inhibitors. Here, we will focus on recent data supporting a role for RhoGDIalpha in the crosstalk between RhoGTPases and present our results suggesting that "preferential" RhoGDIalpha-mediated crosstalk takes place between closely related Rho GTPases.

7.
Cardiovasc Res ; 93(3): 480-9, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22180604

ABSTRACT

AIMS: Although an excessive extracellular matrix remodelling has been well described in myxomatous mitral valve (MMV), the underlying pathogenic mechanisms remain largely unknown. Our goal was to identify dysregulated genes in human MMV and then to evaluate their functional role in the progression of the disease. METHODS AND RESULTS: Dysregulated genes were investigated by transcriptomic, immunohistochemistry, and western blot analyses of the P2 segment collected from human idiopathic MMV during valvuloplasty (n = 23) and from healthy control valves (n = 17). The most striking results showed a decreased expression of two families of genes: the metallothioneins-1 and -2 (MT1/2) and members of the ADAMTS. The mechanistic consequences of the reduced level of MT1/2 were evaluated by silencing their expression in normal valvular interstitial cells (VICs) cultures. The knock-down of MT1/2 resulted in the up-regulation of transforming growth factor-beta 2 (TGF-ß2). Most importantly, TGF-ß2 was also found significantly increased in MMV tissues. The activation of VICs in vitro by TGF-ß2 induced a down-regulation of ADAMTS-1 and an accumulation of versican as observed in human MMV. CONCLUSION: Our studies demonstrate for the first time that MMV are characterized by reduced levels of MT1/2 accompanied by an up-regulation of TGF-ß2. In turn, increased TGF-ß2 signalling induces down-regulation of aggrecanases and up-regulation of versican, two co-operating processes that potentially participate in the development of the pathology.


Subject(s)
Metallothionein/metabolism , Mitral Valve Insufficiency/metabolism , Mitral Valve/metabolism , Transforming Growth Factor beta2/metabolism , Ventricular Remodeling/physiology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Adult , Aged , Cells, Cultured , Female , Humans , Male , Metallothionein/genetics , Microarray Analysis , Middle Aged , Mitral Valve Insufficiency/physiopathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/genetics , Up-Regulation/physiology , Versicans/metabolism
8.
Mol Biol Cell ; 22(17): 3263-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21757538

ABSTRACT

RhoGTPases are key signaling molecules regulating main cellular functions such as migration, proliferation, survival, and gene expression through interactions with various effectors. Within the RhoA-related subclass, RhoA and RhoC contribute to several steps of tumor growth, and the regulation of their expression affects cancer progression. Our aim is to investigate their respective contributions to the acquisition of an invasive phenotype by using models of reduced or forced expression. The silencing of RhoC, but not of RhoA, increased the expression of genes encoding tumor suppressors, such as nonsteroidal anti-inflammatory drug-activated gene 1 (NAG-1), and decreased migration and the anchorage-independent growth in vitro. In vivo, RhoC small interfering RNA (siRhoC) impaired tumor growth. Of interest, the simultaneous knockdown of RhoC and NAG-1 repressed most of the siRhoC-related effects, demonstrating the central role of NAG-1. In addition of being induced by RhoC silencing, NAG-1 was also largely up-regulated in cells overexpressing RhoA. The silencing of RhoGDP dissociation inhibitor α (RhoGDIα) and the overexpression of a RhoA mutant unable to bind RhoGDIα suggested that the effect of RhoC silencing is indirect and results from the up-regulation of the RhoA level through competition for RhoGDIα. This study demonstrates the dynamic balance inside the RhoGTPase network and illustrates its biological relevance in cancer progression.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Gene Expression , Gene Expression Regulation, Neoplastic , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Osteonectin/metabolism , RNA Interference , p38 Mitogen-Activated Protein Kinases/metabolism , rho GTP-Binding Proteins/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Associated Kinases/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein/genetics , rhoC GTP-Binding Protein
9.
J Biol Chem ; 283(31): 21588-98, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18524772

ABSTRACT

RhoA plays a significant role in actin stress fibers formation. However, silencing RhoA alone or RhoA and RhoC did not completely suppress the stress fibers suggesting a residual "Rho-like" activity. RhoB, the third member of the Rho subclass, is a shortlived protein barely detectable in basal conditions. In various cell types, the silencing of RhoA induced a strong up-regulation of both total and active RhoB protein levels that were rescued by re-expressing RhoA and related to an enhanced half-life of the protein. The RhoA-dependent regulation of RhoB does not depend on the activity of RhoA but is mediated by its GDP-bound form. The stabilization of RhoB was not dependent on isoprenoid biosynthesis, Rho kinase, extracellular signal-regulated kinase, p38 mitogen-activated kinase, or phosphatidylinositol 3'-OH kinase pathways but required RhoGDIalpha. The forced expression of RhoGDIalpha increased RhoB half-life, whereas its knock-down antagonized the induction of RhoB following RhoA silencing. Moreover, a RhoA mutant (RhoAR68E) unable to bind RhoGDIalpha was significantly less efficient as compared with wild-type RhoA in reversing RhoB up-regulation upon RhoA silencing. These results suggest that, in basal conditions, RhoGDIalpha is rate-limiting and the suppression of RhoA makes it available to stabilize RhoB. Our results highlight RhoGDIalpha-dependent cross-talks that regulate the stability of RhoGTPases.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanosine Diphosphate/chemistry , rhoA GTP-Binding Protein/physiology , rhoB GTP-Binding Protein/metabolism , Animals , Gene Silencing , Genetic Vectors , Humans , Models, Biological , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection , Up-Regulation , rho GTP-Binding Proteins/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
10.
Free Radic Biol Med ; 44(9): 1732-51, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18325348

ABSTRACT

The role of TGF-beta1 in hydrogen peroxide-induced senescence-like morphogenesis has been described. The aim of this work was to investigate whether TGF-beta1-independent changes in protein synthesis are involved in this morphogenesis and to study possible mechanisms occurring earlier than TGF-beta1 overexpression. Among the multiple TGF-beta1-independent changes in protein neosynthesis, followed or not by posttranslational modifications, identified by proteomic analysis herein, those of ezrin, L-caldesmon, and HSP27 were particularly studied. Rho-GTPase cdc42 was shown to be responsible for p38(MAPK) activation, in turn triggering phosphorylation of L-caldesmon and HSP27. Cdc42 was also shown to be mainly responsible for the increase in TGF-beta1 mRNA level observed at 24 h after treatment with H(2)O(2) and onward. This study further clarified the mechanisms of senescence-like morphogenesis in addition to the previously demonstrated role of TGF-beta1 signaling pathways.


Subject(s)
Fibroblasts/metabolism , Hydrogen Peroxide/pharmacology , Transforming Growth Factor beta1/metabolism , cdc42 GTP-Binding Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Calmodulin-Binding Proteins/pharmacology , Cell Survival , Cellular Senescence , Electrophoresis, Gel, Two-Dimensional , Free Radicals , Humans , Hydrogen Peroxide/chemistry , Models, Biological , Oxidative Stress , Phenotype , Phosphorylation
11.
J Cell Biochem ; 103(3): 857-64, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17615554

ABSTRACT

Rac1 is a Rho subfamily small GTPase which is highly expressed in epidermal keratinocytes. In mice the significance of Rac1 for the maintenance of the epidermis has been controversial. In keratinocytes from human origin, the role of Rac1 in the control of growth/differentiation is still obscure. In this study we used RNA interference to induce specific inhibition of Rac1 expression in cultured human keratinocytes and analyzed the consequences on proliferation and differentiation. We found that the autocrine proliferation of keratinocytes is unaltered by Rac1 silencing. However, the suppression of Rac1 induced premature differentiation as revealed by the expression of markers (keratin 10, involucrin), but the involved mechanism is independent of the activity of p38 mitogen-activated protein kinase. Rather, we found that the effects of Rac1 silencing on keratinocytes differentiation are concomitant with negative regulation of the Ser62/Thr58 phosphorylation on the transcription factor c-myc, a mechanism known to control post-translational stability of the c-myc protein. Thus, in growing human keratinocytes, Rac1 could impede the expression of premature differentiation markers, probably by exerting positive control on c-myc activity and its binding to specific promoters.


Subject(s)
Cell Proliferation , Keratinocytes/cytology , Keratinocytes/metabolism , rac1 GTP-Binding Protein/metabolism , Adult , Animals , Cell Differentiation , Cells, Cultured , Humans , Keratinocytes/physiology , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , Skin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rho GTP-Binding Proteins/metabolism
12.
J Cell Biol ; 179(6): 1261-73, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18086921

ABSTRACT

Ultraviolet B and genotoxic drugs induce the expression of a vascular endothelial growth factor A (VEGF-A) splice variant (VEGF111) encoded by exons 1-4 and 8 in many cultured cells. Although not detected in a series of normal human and mouse tissue, VEGF111 expression is induced in MCF-7 xenografts in nude mice upon treatment by camptothecin. The skipping of exons that contain proteolytic cleavage sites and extracellular matrix-binding domains makes VEGF111 diffusible and resistant to proteolysis. Recombinant VEGF111 activates VEGF receptor 2 (VEGF-R2) and extracellularly regulated kinase 1/2 in human umbilical vascular endothelial cells and porcine aortic endothelial cells expressing VEGF-R2. The mitogenic and chemotactic activity and VEGF111's ability to promote vascular network formation during embyonic stem cell differentiation are similar to those of VEGF121 and 165. Tumors in nude mice formed by HEK293 cells expressing VEGF111 develop a more widespread network of numerous small vessels in the peritumoral tissue than those expressing other isoforms. Its potent angiogenic activity and remarkable resistance to proteolysis makes VEGF111 a potential adverse factor during chemotherapy but a beneficial therapeutic tool for ischemic diseases.


Subject(s)
Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Camptothecin/pharmacology , DNA Damage , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Gene Expression/radiation effects , Glycosylation , Humans , Hypoglycemia/metabolism , Hypoxia/metabolism , Mice , Mice, Nude , Mutagens/pharmacology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Swine/metabolism , Ultraviolet Rays , Vascular Endothelial Growth Factor A/genetics
13.
Biochem Biophys Res Commun ; 359(3): 834-9, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17568564

ABSTRACT

Proliferation of dermal fibroblasts is crucial for the maintenance of skin. The small Rho GTPase, Rac1, has been identified as a key transducer of proliferative signals in various cell types, but in normal human dermal fibroblasts its significance to cell growth control has not been studied. In this study, we applied the method of RNA interference to suppress endogenous Rac1 expression and examined the consequences on human skin fibroblasts. Rac1 knock-down resulted in inhibition of DNA synthesis. This effect was not mediated by inhibition of the central transducer of proliferative stimuli, ERK1/2 or by activation of the pro-apoptotic p38. Rather, as a consequence of the suppressed Rac1 expression we observed a significant decrease in phosphorylation of c-myc, revealing for the first time that in human fibroblasts Rac1 exerts control on proliferation through c-myc phosphorylation. Thus Rac1 activates proliferation of normal fibroblasts through stimulation of c-myc phosphorylation without affecting ERK1/2 activity.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Skin/cytology , Skin/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Small Interfering/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/genetics
14.
J Cell Sci ; 118(Pt 6): 1173-83, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15728253

ABSTRACT

The small GTPases of the Rho family are key intermediates in cellular signalling triggered by activated cell-adhesion receptors. In this study, we took advantage of RNA interference (RNAi) using small interfering RNAs (siRNAs) to define the roles of the best-characterized members of the RhoGTPase family, RhoA, Rac1 and Cdc42, in the control of MMP-1, MMP-2 and type-I-collagen expression in normal human skin fibroblasts (HSFs). A specific and long-lasting repression, up to 7 days after transfection, of the three GTPases was achieved by transient transfection of specific siRNA. The silencing of Cdc42, but not that of RhoA or Rac1, induced a 15-fold increase in MMP-1 secretion. This upregulation was confirmed at the mRNA level and observed with two different siRNAs targeting Cdc42. Such a regulation was also observed in various human cell lines and was rescued by re-expressing wild-type Cdc42 encoded by a construct bearing silent mutations impeding its recognition by the siRNA. By contrast, MMP-2 and type-I-collagen expression was not affected by the individual silencing of each Rho GTPase. Cytokine protein array, enzyme-linked immunosorbent assays and reverse-transcription PCR measurements revealed that ablation of Cdc42 induced an overexpression of interleukin 8 and MCP-1. Although these cytokines are known to induce the expression of MMP-1, we showed that they were not involved in the Cdc42-mediated upregulation of MMP-1. Silencing of Cdc42 also induced an increased phosphorylation of ERK1/2 and p38 MAP kinase. The use of chemical inhibitors on Cdc42-ablated cells revealed that the upregulation of MMP-1 is dependent on the ERK1/2 pathways, whereas the p38 MAP kinase pathway displayed an inhibitory role. Simultaneous knock-down of two or three Rho GTPases allowed us to demonstrate that the RhoA-ROCK pathway was not involved in this regulation but that the silencing of Rac1 reduced the effect of Cdc42 suppression. These data suggest that, in vivo, when cell/extracellular-matrix interactions via integrins induce cytoskeleton organization, MMP-1 expression is maintained at a low level by Cdc42 via a repression of the Rac1 and ERK1/2 pathways. Therefore, Cdc42 contributes to ECM homeostasis and connective tissue integrity.


Subject(s)
Down-Regulation , Matrix Metalloproteinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , cdc42 GTP-Binding Protein/physiology , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Culture Media, Serum-Free/pharmacology , DNA Primers/chemistry , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , GTP Phosphohydrolases/metabolism , Gene Silencing , Humans , Immunoblotting , Interleukin-8/metabolism , Microscopy, Phase-Contrast , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Time Factors , Transfection , Up-Regulation , cdc42 GTP-Binding Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
15.
FASEB J ; 16(13): 1802-4, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12354694

ABSTRACT

Endostatin and angiostatin are known as tumor-derived angiogenesis inhibitors, but their mechanisms of action are not yet completely defined. We report here that endostatin and angiostatin, delivered by adenoviral vectors, reduced in vitro the neovessel formation in the mouse aortic ring assay by 85 and 40%, respectively. We also demonstrated in vivo that both endostatin and angiostatin inhibited local invasion and tumor vascularization of transplanted murine malignant keratinocytes, and reduced by 50 and 90% the development of highly vascularized murine mammary tumors. This inhibition of tumor growth was associated with a reduction of tumor vascularization. Expression analysis of vascular endothelial growth factor (VEGF) carried out in the mouse aortic ring model revealed a 3- to 10-fold down-regulation of VEGF mRNA expression in endostatin-treated rings. A similar down-regulation of VEGF expression at both mRNA and protein levels was also observed in the two in vivo cancer models after treatment with each angiogenesis inhibitor. This suggests that endostatin and angiostatin effects may be mediated, at least in part, by their ability to down-regulate VEGF expression within the tumor. This work provides evidence that endostatin and angiostatin act on tumor cells themselves.


Subject(s)
Collagen/physiology , Endothelial Growth Factors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Peptide Fragments/physiology , Plasminogen/physiology , Adenoviridae/genetics , Angiostatins , Animals , Aorta, Thoracic/metabolism , Blood Vessels/growth & development , Blotting, Western , Collagen/genetics , Culture Techniques , Down-Regulation , Endostatins , Endothelial Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Peptide Fragments/genetics , Plasminogen/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Oncogene ; 21(3): 427-36, 2002 Jan 17.
Article in English | MEDLINE | ID: mdl-11821955

ABSTRACT

Angiogenesis is a complex biological process involving the coordinated modulation of many genes. Histone deacetylases (HDAC) are a growing family of enzymes that mediate the availability of chromatin to the transcriptional machinery. Trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA), two HDAC inhibitors known to relieve gene silencing, were evaluated as potential antiangiogenic agents. TSA and SAHA were shown to prevent vascular endothelial growth factor (VEGF)-stimulated human umbilical cord endothelial cells (HUVEC) from invading a type I collagen gel and forming capillary-like structures. SAHA and TSA inhibited the VEGF-induced formation of a CD31-positive capillary-like network in embryoid bodies and inhibited the VEGF-induced angiogenesis in the CAM assay. TSA also prevented, in a dose-response relationship, the sprouting of capillaries from rat aortic rings. TSA inhibited in a dose-dependent and reversible fashion the VEGF-induced expression of VEGF receptors, VEGFR1, VEGFR2, and neuropilin-1. TSA and SAHA upregulated the expression by HUVEC of semaphorin III, a recently described VEGF competitor, at both mRNA and protein levels. This effect was specific to endothelial cells and was not observed in human fibroblasts neither in vascular smooth muscle cells. These observations provide a conspicuous demonstration that HDAC inhibitors are potent anti-angiogenic factors altering VEGF signaling.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Growth Factors/pharmacology , Histone Deacetylase Inhibitors , Lymphokines/pharmacology , Semaphorin-3A , Signal Transduction/drug effects , Animals , Aorta/drug effects , Blotting, Western , Carrier Proteins/genetics , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal, 28S/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...