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










Publication year range
1.
Article in English | MEDLINE | ID: mdl-26636134

ABSTRACT

BACKGROUND: Secreted Protein Acidic and Rich in Cysteine (SPARC) is a matricellular protein which is implicated in regulation of angiogenesis. PURPOSE: To characterize the changes in SPARC expression and effect of its deletion in a mouse model Oxygen Induced Retinopathy (OIR). MATERIALS AND METHODS: Wild type (wt) and SPARC-deficient mice were subjected to high oxygen (75%) for 5 days (p7-p12) before room air for additional 5 days (p12-p17). Retinas from both groups were flat mounted and retinal vessels were labeled with Isolectin-B4. Areas of Retinal Neovascularization (RNV) and vaso-obliteration were measured by Image-J and normalized to total retinal areas. SPARC expression was analyzed in both groups at p14 and p17 in retinal homogenates and sections by Western Blotting (WB) and immunofluorescence respectively. Human Retinal Endothelial Cells (HRECs) were exposed to hypoxia (1% O2) for 6 hours then SPARC was measured in cell lysate and condition medium by WB and ELISA. Moreover, HRECs were treated with VEGF or SPARC to study their mutual regulatory effect. RESULTS: SPARC-deficient mice demonstrated significant increase in the vaso-obliteration (p=0.03) and modest increase in RNV compared to the wt control. Retinal levels of SPARC was significantly decreased during OIR at p14 (p=0.01) and partially restored to normal level by p17. Moreover, hypoxia significantly reduced SPARC expression and secretion in HRECs (p=0.001). We noticed a mutual positive regulatory feedback between SPARC and VEGF. CONCLUSION: SPARC deletion enhances ischemic retinopathy, thus modulation of SPARC expression could be a novel therapeutic approach to prevent pathological RNV.

2.
Neoplasia ; 14(4): 259-70, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22577342

ABSTRACT

Epithelial ovarian cancer is the fourth leading cause of death from gynecologic malignancies in the United States. Most cases are diagnosed at late stages, with the solid tumor masses growing as peritoneal implants, or floating within the ascitic fluid (peritoneal ovarian carcinomatosis). Despite aggressive surgical "debulking," recurrence of recalcitrant disease is frequent with poor patient survival. Efforts to improve survival rates are hindered by lack of biomarkers that can detect and effectively treat ovarian cancer in its early stages. Urokinase plasminogen activator receptor (uPAR) is a multifunctional receptor involved in a myriad of tumor cell processes. However, the role of host uPAR in ovarian cancer is still elusive. To define the potential proinflammatory role of uPAR in ovarian cancer, first, using a syngeneic murine model in uPAR(-/-) mice, we found that ablation of uPAR restrained tumor take and peritoneal implants and prolonged the survival of uPAR(-/-) mice compared with their uPAR(+/+) counterparts. Ascitic fluid accumulation was significantly decreased in uPAR(-/-) mice with decreased macrophage infiltration. Second, in vitro mechanistic studies revealed that host uPAR is involved in the multiple steps of peritoneal metastatic cascade. Third, we evaluated the prognostic utility of tumor and stromal uPAR in human ovarian cancer tissue microarray. In summary, our studies indicated that uPAR plays a significant role in ovarian cancer cell-stromal crosstalk and contributes to increased vascular permeability and inflammatory ovarian cancer microenvironment. This provides a rationale for targeting the uPAR with either specific neutralizing antibodies or targeting its downstream inflammatory effectors in patients with ovarian cancer.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/metabolism , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Blotting, Western , Capillary Permeability/physiology , Carcinoma/pathology , Cell Adhesion/physiology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Prognosis , Receptor Cross-Talk/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Transfection , Tumor Microenvironment/physiology
3.
Neoplasia ; 11(2): 126-35, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19177197

ABSTRACT

Epigenetic silencing of tumor suppressor genes is a new focus of investigation in the generation and proliferation of carcinomas. Secreted protein acidic and rich in cysteine (SPARC) is reportedly detrimental to the growth of ovarian cancer cells and has been shown to be epigenetically silenced in several cancers. We hypothesized that SPARC is downregulated in ovarian cancer through aberrant promoter hypermethylation. To that end, we analyzed SPARC expression in ovarian cancer cell lines and investigated the methylation status of the Sparc promoter using methylation-specific polymerase chain reaction. Our results show that SPARC mRNA expression is decreased in three (33%) and absent in four (44%) of the nine ovarian cancer cell lines studied, which correlated with hypermethylation of the Sparc promoter. Treatment with the demethylating agent 5-aza-2'-deoxycytidine rescued SPARC mRNA and protein expression. Addition of exogenous SPARC, as well as ectopic expression by an adenoviral vector, resulted in decreased proliferation of ovarian cancer cell lines. Investigation of primary tumors revealed that the Sparc promoter is methylated in 68% of primary ovarian tumors and that the levels of SPARC protein decrease as the disease progresses from low to high grade. Lastly, de novo methylation of Sparc promoter was shown to be mediated by DNA methyltransferase 3a. These results implicate Sparc promoter methylation as an important factor in the genesis and survival of ovarian carcinomas and provide new insights into the potential use of SPARC as a novel biomarker and/or treatment modality for this disease.


Subject(s)
DNA Methylation , Osteonectin/genetics , Ovarian Neoplasms/genetics , Promoter Regions, Genetic , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Proliferation/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methyltransferase 3A , Decitabine , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Osteonectin/drug effects , Ovarian Neoplasms/drug therapy , RNA, Messenger/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Tumor Cells, Cultured
4.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R952-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176890

ABSTRACT

Medial thickening and vascular hypertrophy of resistance arteries can lead to cardiovascular complications associated with diabetes. While previous studies have established a role of type 1 diabetes in vascular remodeling, we recently extended these observations to type 2 diabetes and reported increased collagen deposition due to alterations in matrix metalloproteinase expression and activity in mesenteric resistance arteries. These studies also showed that remodeling response was mediated by endothelin-1 (ET-1) via activation of ET(A) receptors, whereas blockade of ET(B) receptors exacerbated the remodeling. However, the effectiveness of glycemic control strategies in preventing these vascular changes, including activation of the ET system still remained unclear. Also, very little is known about whether and to what extent reorganization of the extracellular matrix (ECM) affects vascular compliance and vasomotor tone. Accordingly, this study assessed structural remodeling of mesenteric microvessels, vascular compliance, and myogenic tone, as well as the role of matrix metalloproteinases (MMP) in mediating these processes. Spontaneously diabetic, non-obese Goto-Kakizaki (GK) rats, a model for type 2 diabetes, and normoglycemic Wistar rats were used for the studies. A subset of GK rats were administered metformin to achieve euglycemia. Glycemic control normalized the increased media-to-lumen ratios (M/L) and myogenic tone seen in diabetes, as well as normalizing plasma ET-1 levels and mesenteric ET(A) receptor expression. There was increased collagen synthesis in diabetes paralleled by decreased collagenase MMP-13 activity, while glycemic control attenuated the process. These findings and our previous study taken together suggest that hyperglycemia-mediated activation of ET-1 and ET(A) receptors alter vascular structure and mechanics in type 2 diabetes.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Endothelin-1/metabolism , Hypoglycemic Agents/pharmacology , Mesenteric Arteries/drug effects , Metformin/pharmacology , Vasoconstriction/drug effects , Animals , Blood Glucose/metabolism , Collagen/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Hyperplasia , Male , Matrix Metalloproteinase 13/metabolism , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Microvessels/drug effects , Microvessels/metabolism , Microvessels/physiopathology , Rats , Rats, Wistar , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism
5.
Neoplasia ; 10(10): 1092-104, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813349

ABSTRACT

We have recently identified that the role of secreted protein acidic and rich in cysteine (SPARC) in amelioration of peritoneal ovarian carcinomatosis is mediated, at least in part, through mesothelial cell/lysophosphatidic acid-induced inflammatory response in ovarian cancer cells. The aim of this study was to elucidate the molecular mechanisms of the interactions between tumor cells and the cellular components of the ovarian cancer peritoneal microenvironment, specifically, mesothelial cells and macrophages. We found that SPARC not only significantly reduced macrophage chemoattractant protein-1 production and its macrophage chemotactic effect, but also attenuated the response of ovarian cancer cells to the mitogenic and proinvasive effects of macrophage chemo-attractant protein-1 and decreased macrophage-induced cancer cell invasiveness. Overexpression of SPARC in ovarian cancer cells significantly attenuated macrophage- and mesothelial cell-induced production and activity of interleukin-6, prostanoids (prostaglandins E2 and 8-isoprostanes) as well as matrix metalloproteinases and urokinase plasminogen activator. Moreover, the effects of SPARC overexpression in ovarian cancer cells were mediated, in part, through inhibition of nuclear factor-kappaB promoter activation. These results indicate, for the first time, that the effects of tumor SPARC as a negative regulator of ovarian cancer are mediated through decreased recruitment of macrophages and downregulation of the associated inflammation.


Subject(s)
Carcinoma/complications , Inflammation/genetics , Osteonectin/physiology , Ovarian Neoplasms/complications , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CCL2/metabolism , Chemokine CCL2/pharmacology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/genetics , Coculture Techniques , Epithelium/metabolism , Epithelium/physiology , Female , Humans , Inflammation/etiology , Macrophages/pathology , Neoplasm Invasiveness , Osteonectin/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , U937 Cells
6.
Mol Cancer Res ; 5(10): 1015-30, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17951402

ABSTRACT

Malignant ascites is a major source of morbidity and mortality in ovarian cancer patients. It functions as a permissive reactive tumor-host microenvironment and provides sustenance for the floating tumor cells through a plethora of survival/metastasis-associated molecules. Using a syngeneic, immunocompetent model of peritoneal ovarian carcinomatosis in SP(-/-) mice, we investigated the molecular mechanisms implicated in the interplay between host secreted protein acidic and rich in cysteine (SPARC) and ascitic fluid prosurvival/prometastasis factors that result in the significantly augmented levels of vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMP). Ascitic fluid-enhanced ID8 invasiveness was mediated through VEGF via a positive feedback loop with MMP-2 and MMP-9 and through activation of alpha(v) and beta(1) integrins. Host SPARC down-regulated the VEGF-MMP axis at the transcriptional and posttranscriptional levels. In vitro, SPARC attenuated the basal as well as VEGF-induced integrin activation in tumor cells. SPARC inhibited the VEGF- and integrin-mediated ID8 proliferation in vitro and significantly suppressed their tumorigenicity in vivo. Relative to SP(+/+), SP(-/-) ascitic fluid contained significantly higher levels of bioactive lipids and exerted stronger chemotactic, proinvasive, and mitogenic effects on ID8 cells in vitro. SP(-/-) ascites also contained high levels of interleukin-6, macrophage chemoattractant protein-1, and 8-isoprostane (prostaglandin F(2)alpha) that were positively correlated with extensive infiltration of SP(-/-) ovarian tumors and ascites with macrophages. In summary, our findings strongly suggest that host SPARC normalizes the microenvironment of ovarian cancer malignant ascites through down-regulation of the VEGF-integrin-MMP axis, decreases the levels and activity of bioactive lipids, and ameliorates downstream inflammation.


Subject(s)
Ascitic Fluid/pathology , Carcinoma/secondary , Osteonectin/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Animals , Ascitic Fluid/chemistry , Ascitic Fluid/metabolism , Carcinoma/metabolism , Cell Adhesion , Cell Proliferation , Cell Survival , Chemokine CCL2/analysis , Dinoprost/analogs & derivatives , Dinoprost/analysis , Female , Inflammation/metabolism , Inflammation/pathology , Integrins/metabolism , Interleukin-6/analysis , Metalloendopeptidases/metabolism , Mice , Mice, Mutant Strains , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Am J Pathol ; 171(4): 1104-12, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17717147

ABSTRACT

The matricellular protein secreted protein acidic and rich in cysteine (SPARC) modulates cell adhesion, proliferation, matrix deposition, and tissue remodeling. SPARC has been shown to regulate the expression of collagen type I and transforming growth factor-beta1 in mesangial cells and to be highly expressed during tubulointerstitial fibrosis in rat angiotensin (ANG) II infusion models. We hypothesized that SPARC is a downstream effector of ANG II and that loss of host SPARC function provides a protective effect on renal damage and fibrosis associated with ANG II hypertension. Our results revealed that cultured primary mesangial cells displayed a concentration-dependent increase in SPARC expression in response to ANG II. After a 14-day chronic infusion of ANG II, hypertensive SPARC-null mice exhibited significantly attenuated levels of urinary and renal indicators of oxidative stress and inflammation and decreased renal perivascular and tubulointerstitial fibrosis relative to wild-type hypertensive controls. Moreover, the observed renal protective changes in SPARC-null mice were found to be independent of blood pressure. These results identify SPARC as an effector of ANG II signaling and suggest an important role for SPARC in mediating ANG II-induced oxidative stress, inflammation, and fibrosis.


Subject(s)
Angiotensin II/metabolism , Hypertension, Renal/pathology , Osteonectin/deficiency , Angiotensin II/pharmacology , Animals , Blood Pressure/genetics , Disease Progression , Fibrosis , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Humans , Hypertension, Renal/chemically induced , Hypertension, Renal/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Mutant Strains , Osteonectin/genetics , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
8.
Am J Pathol ; 170(3): 1054-63, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17322388

ABSTRACT

The matricellular glycoprotein SPARC (secreted protein acidic and rich in cysteine) has been accorded major roles in regulation of cell adhesion and proliferation, as well as tumorigenesis and metastasis. We have recently reported that in addition to its potent antiproliferative and proapoptotic functions, SPARC also abrogates ovarian carcinoma cell adhesion, a key step in peritoneal implantation. However, the underlying molecular mechanism through which SPARC ameliorates peritoneal ovarian carcinomatosis seems to be multifaceted and has yet to be delineated. Herein, we show that SPARC significantly inhibited integrin-mediated ovarian cancer cell adhesion to extracellular matrix proteins, as well as to peritoneal mesothelial cells. This counteradhesive effect of SPARC was shown to be mediated in part through significant attenuation of cell surface expression and clustering of alpha(v)-integrin subunit, alpha(v)beta(3)- and alpha(v)beta(5)-heterodimers, and beta(1)-subunit, albeit to a lesser extent, in ovarian cancer cells. Moreover, SPARC significantly suppressed both anchorage-dependent and -independent activation of AKT and mitogen-acti-vated protein kinase survival signaling pathways in ovarian cancer cells in response to serum and epidermal growth factor stimulation. In summary, we have identified a novel role of SPARC as a negative regulator of both integrin-mediated adhesion and growth factor-stimulated survival signaling pathways in ovarian cancer.


Subject(s)
Cell Adhesion/physiology , Integrins/metabolism , Osteonectin/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Cell Survival , Extracellular Matrix/metabolism , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Invasiveness/physiopathology
9.
Neoplasia ; 9(1): 23-35, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17325741

ABSTRACT

The interplay between peritoneal mesothelial cells and ovarian cancer cells is critical for the initiation and peritoneal dissemination of, and ascites formation in, ovarian cancer. The production of lysophosphatidic acid (LPA) by both peritoneal mesothelial cells and ovarian cancer cells has been shown to promote metastatic phenotype in ovarian cancer. Herein, we report that exogenous addition or ectopic overexpression of the matricellular protein SPARC (secreted protein acidic and rich in cysteine) significantly attenuated LPA-induced proliferation, chemotaxis, and invasion in both highly metastatic SKOV3 and less metastatic OVCAR3 ovarian cancer cell lines. SPARC appears to modulate these functions, at least in part, through the regulation of LPA receptor levels and the attenuation of extracellular signal-regulated kinase (ERK) 1/2 and protein kinase B/AKT signaling. Moreover, our results show that SPARC not only significantly inhibited both basal and LPA-induced interleukin (IL) 6 production in both cell lines but also attenuated IL-6-induced mitogenic, chemotactic, and proinvasive effects, in part, through significant suppression of ERK1/2 and, to a lesser extent, of signal transducers and activators of transcription 3 signaling pathways. Our results strongly suggest that SPARC exerts a dual inhibitory effect on LPA-induced mesothelial-ovarian cancer cell crosstalk through the regulation of both LPA-induced IL-6 production and function. Taken together, our findings underscore the use of SPARC as a potential therapeutic candidate in peritoneal ovarian carcinomatosis.


Subject(s)
Cell Communication/drug effects , Epithelial Cells/pathology , Lysophospholipids/antagonists & inhibitors , Osteonectin/pharmacology , Ovarian Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemotaxis/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , Interleukin-6/metabolism , Neoplasm Invasiveness , RNA, Messenger/analysis , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction
10.
Angiogenesis ; 9(2): 39-51, 2006.
Article in English | MEDLINE | ID: mdl-16607569

ABSTRACT

Angiogenesis, a critical process in both health and disease, is mediated by a number of signaling pathways. Although proangiogenic stimuli, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and the phorbol ester phorbol-12 myristate-13 acetate (PMA) are known to promote blood vessel formation, their downstream targets are ill defined. We sought to investigate the signaling pathways required for vessel assembly by utilizing a three-dimensional collagen matrix in which human umbilical vein endothelial cells (HUVECs) form tubular structures. Our data show that PMA is sufficient for the induction of angiogenesis, and that protein kinase C (PKC) is necessary for this process. Evaluation of PKC isoforms alpha and sigma revealed that these proteins are uniquely regulated. Characterization of an additional PMA target, protein kinase D (PKD) demonstrated that this enzyme becomes phosphorylated in HUVECs, and may therefore be involved in proangiogenic signaling. Further examination of downstream effectors of PKC showed that extracellular signal-regulated kinase (ERK) is critical for angiogenesis, and is accordingly phosphorylated in response to PMA. Surprisingly however, phosphorylation of ERK is independent of PKC activity. In addition, we show that the PKC target sphingosine kinase (SPK) is required for vessel formation. These findings illustrate the complexities of blood vessel formation, and suggest that activators utilize multiple independent pathways to invoke a complete angiogenic response.


Subject(s)
Neovascularization, Physiologic/drug effects , Protein Kinases/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Cell Culture Techniques , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Activation , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Time Factors , Umbilical Veins/cytology
11.
Am J Pathol ; 167(6): 1739-52, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314484

ABSTRACT

The matricellular glycoprotein SPARC (secreted protein acidic and rich in cysteine) possesses multifaceted roles in modulation of cell-matrix interactions, as well as tumor growth and metastasis. To investigate the influence of host-derived SPARC on peritoneal dissemination of ovarian cancer, we established a murine model that faithfully recapitulates advanced human disease by intraperitoneal injection of syngeneic ID8 ovarian cancer cells into SPARC-null and wild-type mice. Compared to wild-type mice, SPARC-null mice showed significantly shorter survival and developed extensive nodular peritoneal dissemination with hemorrhagic ascitic fluid accumulation. Ascitic fluid collected from SPARC-null mice showed significantly augmented levels and activity of vascular endothelial growth factor and gelatinases. Immunohistochemical analysis of tumor nodules from SPARC-null mice revealed higher proliferation and lower apoptosis indices with minimal staining for major extracellular matrix constituents. In vitro, SPARC significantly suppressed adhesion to and invasion of various peritoneal extracellular matrix constituents by murine and human ovarian cancer cell lines. Our findings suggest that SPARC ameliorates ovarian peritoneal carcinomatosis through abrogation of the initial steps of disease pathogenesis, namely tumor cell adhesion and invasion, inhibition of tumor cell proliferation, and induction of apoptosis. Thus, SPARC represents an important therapeutic candidate in ovarian cancer.


Subject(s)
Osteonectin/deficiency , Ovarian Neoplasms/pathology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/prevention & control , Peritoneal Neoplasms/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/analysis
12.
J Cell Biochem ; 91(5): 915-25, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15034927

ABSTRACT

Secreted protein acidic and rich in cysteine (SPARC), a member of the family of matricellular proteins, regulates the interaction of cells with pleiotropic factors and proteins of the extracellular matrix (ECM). Although it has been appreciated that transforming growth factor beta 1 (TGF-beta1) induces SPARC and collagen type I, we have recently shown that SPARC regulates the expression of TGF-beta1 and collagen type I in renal mesangial cells via a TGF-beta1-dependent pathway, and have proposed a reciprocal, autocrine regulatory feedback loop between SPARC and TGF-beta1. Herein, we sought to determine how SPARC regulates TGF-beta1-dependent signal transduction. Our data indicate that SPARC modulates the TGF-beta1-dependent phosphorylation of Smad-2 in primary mesangial cells derived from wild-type and SPARC-null mice. We also show that SPARC regulates the levels and activation of the stress-activated c-jun-N-terminal kinase (JNK) in mesangial cells by augmentation of the stimulatory effects of TGF-beta1. Furthermore, we found that SPARC increases the levels and the activity of the transcription factor c-jun. These effects of SPARC on the TGF-beta1 signaling pathway appear to be mediated through an interaction with the TGF-beta1-receptor complex, but only in the presence of TGF-beta1 bound to its cognate type II receptor. That SPARC is directly involved in the regulation of the TGF-beta1 signaling cascade is consistent with the paradigm that matricellular proteins modulate interactions among cells, growth factors, and their respective receptors.


Subject(s)
Glomerular Mesangium/physiology , Osteonectin/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Animals , Blotting, Western , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Drug Synergism , Glomerular Mesangium/cytology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4 , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Osteonectin/genetics , Osteonectin/pharmacology , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/pharmacology , Smad2 Protein , Sp1 Transcription Factor/metabolism , Trans-Activators/analysis , Trans-Activators/metabolism , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
13.
Angiogenesis ; 6(1): 47-54, 2003.
Article in English | MEDLINE | ID: mdl-14517404

ABSTRACT

Fibroblast growth factor-1 (FGF-1) is a potent angiogenic factor; its structure lacks a signal peptide for secretion. We previously reported that the overexpression of a secreted version of FGF-1 (sp-FGF-1) in microvascular endothelial cells (ECs) enhances cell migration [Partridge et al. J Cell Biochem 2000; 78(3): 487]. In the current study, we have examined the angiogenic effects of sp-FGF-1 in chicken chorioallantoic membranes (CAMs). Two methods of examining the effects of sp-FGF-1 in CAMs were used: cell-mediated transfection via bovine ECs and direct gene transfection. In the cell-mediated gene transfection, those eggs that were implanted with a gelatin sponge seeded with ECs stably transfected to over-express sp-FGF-1 protein showed a significant increase in angiogenesis inside the sponge when compared to eggs treated with vector control-transfected ECs. In the direct gene transfer, eggs received sp-FGF-1 showed a significant increase in vascularization when compared to eggs received vector alone plasmids. These CAM models are useful both for studying molecular mechanisms of angiogenesis and for developing better gene therapy strategies.


Subject(s)
Allantois/metabolism , Chorion/metabolism , Fibroblast Growth Factor 1/metabolism , Neovascularization, Physiologic/genetics , Animals , Cattle , Chick Embryo , Endothelial Cells/metabolism , Fibroblast Growth Factor 1/genetics , Gene Transfer Techniques , Immunohistochemistry , Neovascularization, Physiologic/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
J Cell Biochem ; 90(2): 408-23, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14505356

ABSTRACT

The role of the matricellular protein SPARC (secreted protein, acidic and rich in cysteine) in modulation of vascular cell proliferation is believed to be mediated, in part, by its ability to regulate the activity of certain growth factors through direct binding. In this study, we demonstrate that SPARC does not bind to basic fibroblast growth factor (bFGF/FGF-2) or interfere with complex formation between FGF-2 and its high-affinity FGF receptor-1 (FGFR1), yet both native SPARC and a peptide derived from the C-terminal high-affinity Ca(2+)-binding region of protein significantly inhibit ligand-induced autophosphorylation of FGFR1 (>80%), activation of mitogen-activated protein kinases (MAPKs) (>75%), and DNA synthesis in human microvascular endothelial cells (HMVEC) stimulated by FGF-2 (>80%). We also report that in the presence of FGF-2, a factor which otherwise stimulates myoblast proliferation and the repression of terminal differentiation, both native SPARC and the Ca(2+)-binding SPARC peptide significantly promote (>60%) the differentiation of the MM14 murine myoblast cell line that expresses FGFR1 almost exclusively. Moreover, using heparan sulfate proteoglycan (HSPG)-deficient myeloid cells and porcine aortic endothelial cells (PAECs) expressing chimeric FGFR1, we show that antagonism of FGFR1-mediated DNA synthesis and MAPK activation by SPARC does not require the presence of cell-surface, low-affinity FGF-2 receptors, but can be mediated by an intracellular mechanism that is independent of an interaction with the extracellular ligand-binding domain of FGFR1. We also report that the inhibitory effect of SPARC on DNA synthesis and MAPK activation in endothelial cells is mediated in part (>50%) by activation of protein kinase A (PKA), a known regulator of Raf-MAPK pathway. SPARC thus modulates the mitogenic effect of FGF-2 downstream from FGFR1 by selective regulation of the MAPK signaling cascade.


Subject(s)
Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Osteonectin/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Aorta/metabolism , Calcium/metabolism , Cell Division , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/deficiency , Humans , Ligands , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Peptide Fragments/pharmacology , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1 , Recombinant Proteins/metabolism , Signal Transduction , Swine
15.
J Cell Biochem ; 88(4): 802-11, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12577314

ABSTRACT

Glomerular mesangial cells both synthesize and respond to insulin-like growth factor-1 (IGF-1). Increased activity of the IGF signaling pathway has been implicated as a major contributor to renal enlargement and subsequent development of diabetic nephropathy. Secreted protein acidic and rich in cysteine (SPARC), a matricellular protein, has been shown to modulate the interaction of cells with growth factors and extracellular matrix. We have reported that primary glomerular mesangial cells derived from SPARC-null mice exhibit an accelerated rate of proliferation and produce substantially decreased levels of transforming growth factor beta1 (TGF-beta1) in comparison to their wild-type counterparts (Francki et al. [1999] J. Biol. Chem. 274: 32145-32152). Herein we present evidence that SPARC modulates IGF-dependent signaling in glomerular mesangial cells. SPARC-null mesangial cells produce increased amounts of IGF-1 and -2, as well as IGF-1 receptor (IGF-1R) in comparison to wild-type cells. Addition of recombinant SPARC to SPARC-null cells inhibited IGF-1-stimulated mitogen activated protein kinase (MAPK) activation and DNA synthesis. We also show that the observed accelerated rate of basal and IGF-1-stimulated proliferation in mesangial cells derived from SPARC-null animals is due, at least in part, to markedly diminished levels of cyclin D1 and the cyclin-dependent kinase (cdk) inhibitors p21 and p27. Since expression of SPARC in the glomerulus is especially prominent during renal injury, our findings substantiate previous claims that SPARC is involved in glomerular remodeling and repair, a process commonly associated with mesangioproliferative glomerulonephritis and diabetic nephropathy.


Subject(s)
Glomerular Mesangium/physiology , Osteonectin/physiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/biosynthesis , Cell Division/drug effects , Cells, Cultured , Diabetic Nephropathies/physiopathology , Glomerular Mesangium/drug effects , Glomerulonephritis/physiopathology , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/biosynthesis , Mice , Osteonectin/deficiency , Osteonectin/pharmacology , Protein Kinase Inhibitors , Protein Kinases/biosynthesis , RNA/analysis , RNA/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptor, IGF Type 1/biosynthesis , Signal Transduction/drug effects , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta1
16.
J Cell Biochem ; 84(4): 759-71, 2002.
Article in English | MEDLINE | ID: mdl-11835401

ABSTRACT

Interactions among growth factors, cells, and extracellular matrix regulate proliferation during normal development and in pathologies such as atherosclerosis. SPARC (secreted protein, acidic, and rich in cysteine) is a matrix-associated glycoprotein that modulates the adhesion and proliferation of vascular cells. In this study, we demonstrate that SPARC inhibits human arterial smooth muscle cell proliferation stimulated by platelet-derived growth factor or by adhesion to monomeric type I collagen. Binding studies with SPARC and SPARC peptides indicate specific and saturable interaction with smooth muscle cells that involves the C-terminal Ca2+-binding region of the protein. We also report that SPARC arrests monomeric collagen-supported smooth muscle cell proliferation in the late G1-phase of the cell cycle in the absence of an effect on cell shape or on levels of cyclin-dependent kinase inhibitors. Cyclin-dependent kinase-2 activity, p107 and cyclin A levels, and retinoblastoma protein phosphorylation are markedly reduced in response to the addition of exogenous SPARC and/or peptides derived from specific domains of SPARC. Thus, SPARC, previously characterized as an inhibitor of platelet-derived growth factor binding to its receptor, also antagonizes smooth muscle cell proliferation mediated by monomeric collagen at the level of cyclin-dependent kinase-2 activity.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , G1 Phase/drug effects , Muscle, Smooth, Vascular/drug effects , Osteonectin/pharmacology , Platelet-Derived Growth Factor/pharmacology , Amino Acid Sequence , Animals , Aorta/cytology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/physiology , Cell Size/drug effects , Cell Size/physiology , Collagen Type I/metabolism , Drug Interactions , Extracellular Matrix/metabolism , G1 Phase/physiology , Humans , Mice , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Osteonectin/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...