Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Endocrinol Metab ; 85(7): 2609-14, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10902815

ABSTRACT

One of the initial stages of adipogenesis is migration of preadipocytes of mesenchymal origin into cell clusters to form primitive fat organs. The serine protease inhibitor plasminogen activator inhibitor-1 (PAI-1) is synthesized and released from human adipose tissue ex vivo and regulates smooth muscle and endothelial cell migration in vitro, but its role in adipose tissue is not known. We investigated the role of PAI-1 in cultures of human preadipocytes from men and women of various ages and body mass indexes. Human preadipocytes expressed the messenger ribonucleic acid for PAI-1 and released significant quantities of PAI-1 protein into the medium. As PAI-1 regulates motility through the interaction of vitronectin with its receptor, the integrin alphaVbeta3, we identified this receptor in human preadipocytes. Flow cytometric analysis indicated that human preadipocytes express the vitronectin receptor alphaVbeta3 in a similar pattern as human umbilical vein endothelial cells. Functional studies indicated that active, but not latent, PAI-1 inhibited preadipocyte attachment to vitronectin with an IC(50) of 13.3 nmol/L, and preincubation of vitronectin-coated Transwells with active PAI-1 prevented preadipocyte migration. Vitronectin was identified in homogenates of the stromal-vascular fraction of human adipose tissue, but was absent from human adipocytes and cultured preadipocytes. These data indicate that human preadipocyte migration is regulated through the endogenous expression of PAI-1 and alphaVbeta3 integrin, a novel autocrine mechanism for potentially regulating cell cluster formation in adipogenesis.


Subject(s)
Adipocytes/physiology , Autocrine Communication/physiology , Plasminogen Activator Inhibitor 1/pharmacology , Serine Proteinase Inhibitors/pharmacology , Adipocytes/drug effects , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adult , Autocrine Communication/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Reverse Transcriptase Polymerase Chain Reaction
2.
Biochem Biophys Res Commun ; 279(3): 984-8, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11162462

ABSTRACT

The effect of insulin and insulin-like growth factor-1 (IGF-1) on plasminogen activator inhibitor-1 (PAI-1) release from primary cultures of human preadipocytes and adipocytes has been investigated. Initial experiments measuring basal PAI-1 release (ng/ml) indicated variability between individual cultures. Using a novel technique for adipocyte quantitation, additional experiments were performed to determine PAI-1 release per cell, indicating a significant reduction with differentiation. Insulin and IGF-1 over a range of concentrations had no effect on PAI-1 release, and RT-PCR of PAI-1 mRNA following treatment with insulin and IGF-1 also indicated similar expression between treatments. The cultures did exhibit insulin-stimulated glyceraldehyde-3-phosphate dehydrogenase (GAPDH) expression and leptin synthesis following differentiation to the adipocyte phenotype. This is the first report of PAI-1 secretion by primary cultures of human preadipocytes and adipocytes, indicating PAI-1 release independent of insulin and IGF-1 and implicating other factors in the elevated plasma PAI-1 observed with insulin resistance.


Subject(s)
Adipocytes/drug effects , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Cells, Cultured , Humans , Stem Cells/drug effects , Stem Cells/metabolism
3.
J Clin Endocrinol Metab ; 84(9): 3222-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487691

ABSTRACT

To further investigate the role of plasminogen activator inhibitor-1 (PAI-1) in adipose tissue physiology, the production and regulation of PAI-1 was determined in primary cultures of human preadipocytes. When expressed as production per cell and cultured under identical conditions, human preadipocytes from both visceral (omental) and sc depots of lean and obese individuals released significant, yet similar, amounts of PAI-1 protein into the conditioned medium. High steady-state PAI-1 messenger RNA (mRNA) concentrations were observed in visceral and sc preadipocytes, with the relative level of expression equivalent to beta-actin mRNA. Tumor necrosis factor alpha significantly decreased PAI-1 production in a concentration-dependent manner in both visceral and sc cultures, whereas transforming growth factor beta significantly elevated PAI-1 production, but only in sc preadipocytes from obese individuals. Addition of insulin had no effect on antigen levels in conditioned medium of preadipocyte cultures. Stimulation of the preadipocyte cultures with a defined medium resulted in differentiation to the adipocyte phenotype, as determined by flow cytometric analysis, verifying the cultures as human preadipocyte. These studies are the first to observe significant PAI-1 mRNA expression and protein production in primary cultures of a human adipose tissue cellular component, and they suggest that nascent adipocytes contribute significantly to the elevated plasma PAI-1 observed in obesity.


Subject(s)
Adipocytes/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/metabolism , Stem Cells/metabolism , Actins/genetics , Adipocytes/drug effects , Cell Count , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned , Gene Expression , Humans , Insulin/pharmacology , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/analysis , Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
4.
Endocrinology ; 140(1): 154-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9886820

ABSTRACT

The role of angiotensin II (AII) in human preadipocyte physiology has been investigated in primary cultures from human adipose tissue. Receptor binding studies indicated that human preadipocytes express a high affinity AII binding site of the AT1 subtype, as binding of 125I-labeled [Sar1,Ile8]AII was rapid, saturable, and specific. As AII has previously been demonstrated to affect the cell cycle in adrenal and cardiac cells, the effect of AII on regulation of cycle progression was examined in human preadipocytes. Stimulation of preadipocytes with AII resulted in G1 phase progression of the cell cycle, as determined by flow cytometric analysis. AII treatment was associated with induction of expression of the messenger RNA for the cell cycle regulatory protein cyclin D1 in a dose-dependent manner. Pretreatment of cells with subtype-selective AT receptor ligands before AII stimulation indicated that the cyclin response was mediated via the AT1 receptor. The identity of the cells as preadipocyte was verified by culture in a defined differentiation medium, observing both leptin message expression and triglyceride accumulation by flow cytometry. These findings indicate that AII has early, receptor-mediated effects on cell cycle progression in human preadipocytes that may contribute to differentiation to the adipocyte phenotype.


Subject(s)
Adipocytes/physiology , Adipose Tissue/cytology , Cell Cycle/physiology , Receptors, Angiotensin/physiology , 1-Sarcosine-8-Isoleucine Angiotensin II/metabolism , Adipocytes/cytology , Adult , Binding Sites , Cell Differentiation , Cell Separation , Cells, Cultured , Cyclin D1/biosynthesis , Cyclin D1/genetics , Female , Flow Cytometry , Humans , Male , RNA, Messenger/metabolism , Receptors, Angiotensin/metabolism
5.
Biochem Biophys Res Commun ; 248(3): 523-6, 1998 Jul 30.
Article in English | MEDLINE | ID: mdl-9703958

ABSTRACT

This study reports the initial observation of the novel estrogen receptor, ER beta, in human subcutaneous adipose tissue. Human adipose tissue was obtained from various anatomic sites including the subcutaneous depots of lipectomy patients (healthy controls), and from subcutaneous abdominal and breast depots of lean and obese women with breast cancer. ER beta mRNA expression, determined by reverse transcription and polymerase chain reaction (RT-PCR), was present in each adipose tissue sample examined. Cloning and sequencing of the PCR product confirmed its identity as ER beta. Separation of adipose tissue into component fractions indicated that ER beta was expressed in both adipocytes and the stroma-vasculature. Primary culture of human preadipocytes indicated that ER beta mRNA was present only after differentiation to the adipocyte phenotype. This novel observation of ER beta mRNA indicates that this receptor subtype may have a role in ER-mediated responses in human adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Receptors, Estrogen/biosynthesis , Transcription, Genetic , Abdomen , Adipose Tissue/pathology , Adult , Aged , Body Mass Index , Body Weight , Breast , Breast Neoplasms/pathology , Cloning, Molecular , Estrogen Receptor beta , Female , Humans , Menopause , Middle Aged , Obesity/metabolism , Polymerase Chain Reaction , Skin , Tumor Cells, Cultured
6.
Exp Cell Res ; 238(2): 324-34, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9473340

ABSTRACT

Various cell adhesion molecules mediate the diverse functions of the vascular endothelium, such as cell adhesion, neutrophil migration, and angiogenesis. In order to identify cell adhesion molecules important for angiogenesis, we used an in vitro model (Chalupowicz, Chowdhury, Bach, Barsigian, and Martinez, J. Cell Biol. 130, 207-215, 1995) in which human umbilical vein endothelial cell monolayers are induced to form capillary-like tubes when a second gel, composed of either fibrin or collagen, is formed overlying the apical surface. In the present investigation, we observed that a monoclonal antibody directed against the first extracellular domain of human vascular endothelial cadherin (VE-cadherin, cadherin 5) inhibited the formation of capillary tubes formed between either fibrin or collagen gels. Moreover, when added to preformed capillary tubes, this antibody disrupted the capillary network. In contrast, monoclonal antibodies directed against the extracellular domain of N-cadherin, the alphavbeta3 integrin, and PECAM-1 failed to inhibit capillary tube formation. During capillary tube formation, Western blot and RT-PCR analysis revealed no marked change in VE-cadherin expression. Immunocytochemical studies demonstrated that VE-cadherin was concentrated at intercellular junctions in multicellular capillary tubes. Thus, VE-cadherin plays a specific role in fibrin-induced or collagen-induced capillary tube formation and is localized at areas of intercellular contact where it functions to maintain the tubular architecture. Moreover, its function at tubular intercellular junctions is distinct from that at intercellular junctions present in confluent monolayers, since only the former was inhibited by monoclonal antibodies.


Subject(s)
Cadherins/physiology , Capillaries/growth & development , Endothelium, Vascular/growth & development , Fibrin , Neovascularization, Physiologic/physiology , Animals , Antibody Specificity , Antigens, CD , Cadherins/analysis , Cadherins/genetics , Cell Division , Cells, Cultured , Collagen , Gels , Humans , Intercellular Junctions/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/physiology , RNA, Messenger/analysis , Rats , Receptors, Vitronectin/physiology , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...