Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Osteoarthritis Cartilage ; 17(8): 1049-55, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19285161

ABSTRACT

OBJECTIVE: Human osteoarthritic (OA) cartilage type-II collagen is preferentially cleaved by the proinflammatory cytokine-induced matrix metalloproteinases-13 (MMP-13). Interferon-gamma (IFN-gamma) potently inhibits interleukin-1 (IL-1)-induced MMP-13 expression in healthy chondrocytes. Our goal was to study the previously unknown impact of IFN-gamma on MMP-13 in OA and compare the levels and functional activity of IFN-gamma receptor (IFN-gammaR1) in healthy and OA chondrocytes. METHODS: Chondrocytes were obtained from OA patients and non-arthritic control subjects and treated with IL-1+ or- IFN-gamma. MMP-13 mRNA and protein expression were measured by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting. IFN-gammaR1 expression was assessed by flow cytometry, immunoprecipitation and immunohistochemistry with fluorescein-labeled antibody. IFN-gammaR1 was neutralized with its antibody and signal transducer and activator of transcription 1 (STAT1) phosphorylation analyzed by Western blotting. OA chondrocytes were also transfected with control and IFN-gammaR1 expression vectors. RESULTS: OA chondrocytes displayed a drastically impaired MMP-13 suppression by IFN-gamma compared to control cells. IFN-gammaR1 levels were significantly decreased in OA chondrocytes as assessed by flow cytometry, immunoprecipitation and immunohistochemistry. Consequently, IFN-gamma-stimulated STAT1 phosphorylation mediated by IFN-gammaR1 was also considerably reduced in OA patient chondrocytes. IFN-gammaR1 overexpression in OA cells restored MMP-13 suppression by IFN-gamma. CONCLUSIONS: Ability of IFN-gamma to suppress IL-1-induced MMP-13 expression is diminished in OA chondrocytes due to decreased IFN-gammaR1 levels, activity and impaired downstream signal transduction. Therefore, IFN-gammaR1 modulation and weakened endogenous IFN-gamma response may be important mechanisms in OA pathogenesis and cartilage degradation.


Subject(s)
Collagen Type II/metabolism , Interferon-gamma/metabolism , Osteoarthritis/pathology , Receptors, Interferon/metabolism , Cells, Cultured , Chondrocytes/metabolism , Female , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 13/metabolism , Middle Aged , Interferon gamma Receptor
2.
Ann Rheum Dis ; 66(6): 782-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17179173

ABSTRACT

BACKGROUND: Despite well-documented immunomodulation by interferon gamma (IFNgamma), its role and mechanism of regulation of matrix metalloproteinase 13 (MMP13) gene expression in human chondrocytes is unknown. OBJECTIVE: To investigate the ability and mechanism of IFNgamma to suppress interleukin 1 (IL1)-induced MMP13 expression in articular chondrocytes. METHODS: Human chondrocytes were treated with IFNgamma or IL1beta alone or in combination. MMP13 mRNA was analysed by semiquantitative reverse transcriptase-PCR. MMP13 protein, phospho-signal transducer and activator of transcription 1 (STAT1) and p44/42 mitogen-activated protein kinase levels were measured by western blotting. MMP13 promoter luciferase, cytomegalovirus cyclic AMP response element-binding protein (CBP)/p300 plasmids and STAT1 small interfering RNA (siRNA) were transfected by the calcium phosphate method. IFNgamma receptor was also neutralised. Activator protein (AP) 1 activity was monitored by the TransAM transcription factor kit. STAT1-CBP/p300 interaction was studied by immunoprecipitation. RESULTS: IFNgamma potently suppressed IL1-induced expression of MMP13 and promoter activity. Blockade with neutralising IFNgamma R1 antibody revealed that MMP13 inhibition by IFNgamma is mediated by the IFN receptor. IFNgamma-stimulated activation of STAT1 was directly correlated with MMP13 suppression. Knockdown of the STAT1 gene by specific siRNA or its inhibition with fludarabine partially restored the IL1beta induction of MMP13 expression and promoter activity. IFNgamma did not alter AP1 binding ability but promoted physical interaction of STAT1 and CBP/p300 coactivator. p300 overexpression reversed IFNgamma inhibition of endogenous MMP13 mRNA expression and exogenous MMP13 promoter activity. CONCLUSION: IFNgamma, through its receptor, activates STAT1, which binds with CBP/p300 coactivator, sequesters it from the cell system, and thus inhibits transcriptional induction of the MMP13 gene in chondrocytes. IFNgamma and its signalling pathways could be targeted therapeutically for diminishing IL1-induced cartilage degradation by MMP13 in patients with arthritis.


Subject(s)
Chondrocytes/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Interferon-gamma/pharmacology , Interleukin-1beta/antagonists & inhibitors , Matrix Metalloproteinase 13/metabolism , Blotting, Western/methods , Cartilage, Articular/cytology , Cartilage, Articular/enzymology , Chondrocytes/enzymology , E1A-Associated p300 Protein/metabolism , Humans , Interleukin-1beta/pharmacology , Knee Joint/cytology , Knee Joint/enzymology , Matrix Metalloproteinase 13/genetics , Peptides/metabolism , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , Receptors, Interferon/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , STAT1 Transcription Factor/metabolism , Transfection , Interferon gamma Receptor
3.
Am J Physiol Heart Circ Physiol ; 281(1): H30-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406465

ABSTRACT

Angiotensin II-induced growth signaling mechanisms were investigated in vascular smooth muscle cells (VSMCs) from mesenteric arteries of spontaneously hypertensive (SHR) and Wistar-Kyoto rats (WKY). In WKY, angiotensin II significantly increased protein synthesis ([(3)H]leucine incorporation) but not DNA synthesis ([(3)H]thymidine incorporation). In SHR, angiotensin II increased protein and DNA synthesis. VSMCs from both strains expressed angiotensin type 1 (AT(1)) and type 2 (AT(2)) receptors. Losartan (an AT(1) receptor antagonist) but not PD-123319 (an AT(2) receptor antagonist) attenuated angiotensin II-stimulated protein synthesis in WKY VSMCs. In SHR, losartan and PD-123319 partially inhibited angiotensin II-induced VSMC proliferation. The mitogen-activated protein kinase or extracellular signal-regulated protein kinase (ERK) kinase inhibitor PD-98059 blocked VSMC growth responses to angiotensin II in both strains. Angiotensin II increased ERK1/2 activation more in SHR than WKY, an effect inhibited by losartan but not PD-123319. LY-294002 [a phosphatidylinositol-3 (PI3) kinase inhibitor] blocked angiotensin II-stimulated ERK1/2 activation in SHR but not in WKY, whereas bisindolylmaleimide [a protein kinase C (PKC) inhibitor] was ineffective. In conclusion, angiotensin II stimulates VSMC proliferation via AT(1) and AT(2) receptors in SHR. In WKY, angiotensin II induces VSMC hypertrophy via AT(1) receptors. ERK1/2-dependent pathways regulated by intracellular Ca(2+) but not PKC mediate these effects. In SHR VSMCs, PI3 kinase plays a role in augmented angiotensin II-induced ERK1/2 phosphorylation. These angiotensin II-mediated signaling events could contribute to vascular remodeling in SHR.


Subject(s)
Angiotensin II/pharmacology , Hypertension/pathology , Mesenteric Arteries/pathology , Muscle, Smooth, Vascular/pathology , Rats, Inbred SHR/anatomy & histology , Animals , Calcium/physiology , Cell Division/drug effects , Cells, Cultured , Enzyme Activation , Intracellular Membranes/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Kinase C/physiology , Rats , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Receptors, Angiotensin/physiology , Reference Values
4.
J Hypertens ; 19(3 Pt 2): 553-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11327629

ABSTRACT

OBJECTIVES: The present study investigates effects of angiotensin II on activation of extracellular signal-regulated protein kinase (ERK) 1/2, p38 mitogen activated-protein kinase (p38MAPK) and c-Jun amino terminal kinase (JNK) in vascular smooth muscle cells from spontaneously hypertensive rats (SHR). METHODS: Vascular smooth muscle cells (VSMC) from mesenteric arteries of Wistar-Kyoto (WKY) rats and SHR were studied. Angiotensin II-induced phosphorylation of ERK1/2, JNK and p38MAPK were assessed by Western blot analysis. c-fos mRNA expression by angiotensin II was determined by reverse transcriptase-polymerase chain reaction in the absence and presence of PD98059, selective inhibitor of ERK1/2-dependent pathways and SB202190, selective p38MAPK inhibitor. RESULTS: Angiotensin II increased phosphorylation of ERK1/2 and p38MAPK, but not JNK. Responses were significantly increased in SHR compared with WKY. Irbesartan, AT1 receptor antagonist, but not PD123319, AT2 receptor blocker, abolished angiotensin II-induced effects. PP2, selective Src inhibitor, decreased angiotensin II-mediated activation of MAP kinases. Angiotensin II increased c-fos mRNA expression in SHR and had a small stimulatory effect in WKY. These actions were inhibited by PD98059, whereas SB202190 had no effect. CONCLUSIONS: Angiotensin II-induced activation of vascular ERK1/2 and p38MAPK is increased in SHR. These effects are mediated via AT1 receptors, which activate Src-dependent pathways. Overexpression of c-fos mRNA in SHR is due to ERK1/2-dependent, p38MAPK-independent pathways. Our results suggest that angiotensin II activates numerous MAP kinases in VSMCs and that differential activation of these kinases may be important in altered growth signaling in VSMCs from SHR.


Subject(s)
Hypertension/enzymology , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Rats, Inbred SHR/metabolism , Receptors, Angiotensin/physiology , Angiotensin II/pharmacology , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hypertension/pathology , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/pathology , Proto-Oncogene Proteins c-fos/genetics , Pyridines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Reference Values , Vasoconstrictor Agents/pharmacology , p38 Mitogen-Activated Protein Kinases
5.
Hypertension ; 37(2 Pt 2): 574-80, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230337

ABSTRACT

Vascular remodeling in hypertension is associated with cell growth and increased deposition of extracellular matrix components, particularly collagen. Mechanisms underlying these processes are unclear, but MAP kinases, particularly ERK1/2 and p38 MAP kinase, may be important. We studied the role of ERK1/2 and p38 MAP kinase in vascular smooth muscle cell (VSMC) collagen synthesis and growth mediated by angiotensin (Ang) II in spontaneously hypertensive rats (SHR). Cultured mesenteric VSMC from Wistar-Kyoto rats and SHR were used. Phosphorylation of ERK1/2 and p38 MAP kinase were assessed by Western blots with phosphospecific antibodies. Ang II-stimulated DNA and collagen synthesis were determined by measuring incorporation of (3)H-thymidine and (3)H-proline, respectively. mRNA expression of procollagen I and III was determined by reverse transcription-polymerase chain reaction. Ang II increased ERK1/2 and p38 MAP kinase phosphorylation. Responses were augmented in SHR. Effects were inhibited by irbesartan, a selective AT(1) antagonist, but not by PD123319, a selective AT(2) blocker. Ang II stimulated (3)H-thymidine and (3)H-proline incorporation. These actions were enhanced 2- to 3-fold in SHR. PD98059, selective inhibitor of the ERK1/2 pathway, attenuated Ang II-induced growth and collagen effects and normalized responses in SHR. SB212190, a selective p38 MAP kinase inhibitor, did not alter Ang II-elicited DNA synthesis but reduced collagen production and mRNA expression of procollagen I and III in SHR. These data demonstrate that (1) Ang II-mediated activation of p38 and ERK1/2 is increased in SHR, (2) augmented growth responses are generated by ERK1/2-dependent, p38 MAP kinase-independent pathways, and (3) p38 MAP kinase influences Ang II-induced collagen production in SHR but not in Wistar-Kyoto rats. These results indicate differential roles of ERK1/2 and p38 MAP kinase in AT(1)-stimulated VSMC growth and collagen production, which may contribute to vascular remodeling in hypertension.


Subject(s)
Angiotensin II/pharmacology , Collagen/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Angiotensin Receptor Antagonists , Animals , Biphenyl Compounds/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Hypertension/physiopathology , Imidazoles/pharmacology , Irbesartan , Male , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Procollagen/biosynthesis , Proline/metabolism , Pyridines/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Tetrazoles/pharmacology , Thymidine/metabolism , p38 Mitogen-Activated Protein Kinases
6.
Hypertension ; 37(2 Pt 2): 604-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230342

ABSTRACT

Angiotensin II is an important modulator of cell growth through AT(1) receptors, as demonstrated both in vivo and in vitro. We investigated the role of proteins involved in the cell cycle, including cyclin D1, cyclin-dependent kinase 4 (cdk4), and cyclin-dependent kinase inhibitors p21 and p27 in blood vessels of angiotensin II-infused rats and the effect therein of the AT(1)-receptor antagonist losartan. Male Sprague-Dawley rats were infused for 7 days with angiotensin II (120 ng/kg per minute SC) and/or treated with losartan (10 mg/kg per day orally). DNA synthesis in mesenteric arteries was evaluated by radiolabeled (3)H-thymidine incorporation. The expression of cyclin D1, cdk4, p21, and p27, which play critical roles during the G(1)-phase of the cell cycle process, was examined by Western blot analysis. Tail-cuff systolic blood pressure (mm Hg) was elevated (P<0.01, n=9) in angiotensin II-infused rats (161.3+/-8.2) versus control rats (110.1+/-5.3) and normalized by losartan (104.4+/-3.2). Radiolabeled (3)H-thymidine incorporation (cpm/100 microgram DNA) showed that angiotensin II infusion significantly increased DNA synthesis (152+/-5% versus 102+/-6% of control rats, P<0.05). Expression of cyclin D1 and cdk4 was significantly increased in the angiotensin II group to 213.7+/-8% and 263.6+/-37% of control animals, respectively, whereas expression of p21 and p27 was significantly decreased in the angiotensin II group to 23.2+/-10.4% and 10.3+/-5.3% of control animals, respectively. These effects induced by angiotensin II were normalized in the presence of losartan. Thus, when AT(1) receptors are stimulated in vivo, DNA synthesis is enhanced in blood vessels by activation of cyclin D1 and cdk4. Reduction in cell cycle kinase inhibitors p21 and p27 may contribute to activation of growth induced by in vivo AT(1) receptor stimulation.


Subject(s)
Angiotensin II/pharmacology , Blood Vessels/drug effects , Cell Cycle Proteins/metabolism , Receptors, Angiotensin/metabolism , Angiotensin II/administration & dosage , Angiotensin Receptor Antagonists , Animals , Blood Pressure/drug effects , Blood Vessels/metabolism , Body Weight , Cell Division/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA/biosynthesis , Losartan/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Splanchnic Circulation
7.
Life Sci ; 67(13): 1613-23, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10983855

ABSTRACT

The nitric oxide synthase (NOS) inhibitor L-NAME may have growth inhibitory effects in vivo. We investigated in vitro the potential growth inhibitory effects of three different NOS inhibitors: L-NAME (1 mM), LNMMA (1 mM) and aminoguanidine (0.5 mM), on fetal bovine serum (FBS) and platelet derived growth factor (PDGF-BB)-stimulated growth in cultured vascular smooth muscle cells (VSMCs). [3H]-thymidine incorporation into rat mesenteric VSMCs was measured as an index of VSMCs proliferation (DNA synthesis) and activation of extracellular signal regulated kinase (ERK1/2), a major signaling event in cell growth, was measured by western blot assay. PDGF-BB (0-5 ng/mL) and FBS (0-5%) increased [3H]-thymidine incorporation in a dose-dependent manner up to 6-10 fold. L-NAME significantly reduced PDGF-BB (5 ng/ml) and FBS (5%) stimulated DNA synthesis by 46% and 38% respectively. The increase of [3H]-thymidine incorporation induced by PDGF-BB and FBS was unaltered by L-NMMA. In contrast, aminoguanidine induced an increase in FBS and PDGF-BB-stimulated [3H]-thymidine incorporation of 64% and 34% respectively above cells not exposed to aminoguanidine. ERK1/2 phosphorylation induced by PDGF-BB and FBS was not affected by pre-treatment with L-NAME or aminoguanidine. In conclusion, NOS inhibitors differentially influence DNA synthesis in VSMCs: L-NAME inhibits FBS and PDGF-BB-stimulated cellular proliferation whereas aminoguanidine accentuates FBS and PDGF-BB-stimulated VSMCs proliferation. These phenomena are independent of the ERK1/2 pathway. The growth inhibitory effects of L-NAME may be related to differences in properties from other NOS inhibitors, and independent of its ability to inhibit NOS.


Subject(s)
Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Muscle, Smooth, Vascular/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Animals , Arginine/deficiency , Becaplermin , Cell Division/drug effects , Culture Media , DNA/biosynthesis , Drug Interactions , Guanidines/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Phosphorylation/drug effects , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Thymidine/metabolism , omega-N-Methylarginine/pharmacology
8.
Circ Res ; 84(5): 505-15, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10082472

ABSTRACT

This study investigates the role of extracellular signal-regulated kinases (ERKs) in angiotensin II (Ang II)-generated intracellular second messengers (cytosolic free Ca2+ concentration, ie, [Ca2+]i, and pHi) and in contraction in isolated vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR) and control Wistar Kyoto rats (WKY) using the selective mitogen-activated protein (MAP)/ERK inhibitor, PD98059. VSMCs from mesenteric arteries were cultured on Matrigel basement membrane matrix. These cells, which exhibit a contractile phenotype, were used to measure [Ca2+]i, pHi, and contractile responses to Ang II (10(-12) to 10(-6) mol/L) in the absence and presence of PD98059 (10(-5) mol/L). [Ca2+]i and pHi were measured by fura-2 and BCECF methodology, respectively, and contraction was determined by photomicroscopy. Ang II-stimulated ERK activity was measured by Western blot analysis using a phospho-specific ERK-1/ERK-2 antibody and by an MAPK enzyme assay. Ang II increased [Ca2+]i and pHi and contracted cells in a dose-dependent manner. Maximum Ang II-elicited contraction was greater (P<0.05) in SHR (41.9+/-5.1% reduction in cell length relative to basal length) than in WKY (28.1+/-3.0% reduction in cell length relative to basal length). Basal [Ca2+]i, but not basal pHi, was higher in SHR compared with WKY. [Ca2+]i and pHi effects of Ang II were enhanced (P<0.05) in SHR compared with WKY (maximum Ang II-induced response [Emax] of [Ca2+]i, 576+/-24 versus 413+/-43 nmol/L; Emax of pHi, 7.33+/-0.01 versus 7.27+/-0.03, SHR versus WKY). PD98059 decreased the magnitude of contraction and attenuated the augmented Ang II-elicited contractile responses in SHR (Emax,19. 3+/-3% reduction in cell length relative to basal length). Ang II-stimulated [Ca2+]i (Emax, 294+/-55 nmol/L) and pHi (Emax, 7. 27+/-0.04) effects were significantly reduced by PD98059 in SHR. Ang II-induced ERK activity was significantly greater (P<0.05) in SHR than in WKY. In conclusion, Ang II-stimulated signal transduction and associated VSMC contraction are enhanced in SHR. MAP/ERK inhibition abrogated sustained contraction and normalized Ang II effects in SHR. These data suggest that ERK-dependent signaling pathways influence contraction and that they play a role in vascular hyperresponsiveness in SHR.


Subject(s)
Angiotensin II/physiology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Angiotensin II/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hydrogen-Ion Concentration , Male , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Muscle Contraction , Muscle, Smooth, Vascular/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstrictor Agents/pharmacology
9.
Placenta ; 17(7): 471-7, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8899876

ABSTRACT

In human placental syncytiotrophoblast brush-border (BBM, facing the mother) and basal-plasma membranes (BPM, facing to fetus) we have recently demonstrated the presence of calcaemic hormone-specific receptors for parathyroid hormone and calcitonin, which could be implicated in calcium transport from the mother to the fetus. It is well recognized that signal transducing G proteins (guanosinc nucleotide-binding proteins) can associate with various transmembrane receptors and effector proteins, and regulate a variety of second-messenger systems and ion channels. In this present paper, we investigated the presence of a variety of alpha and beta subunits of G proteins in both syncytiotrophoblast, BBM and BPM by Western blot technique. For the first time, we were able to demonstrate the presence of G proteins in the bipolar syncytiotrophoblast membranes, which were evaluated by immunoblotting using affinity purified antiserum raised against the alpha subunits of Gi1, Gi1/i2, Gi3, G0, Gq, Gs, G7 and against the beta subunits. In BBM, we identified the alpha subunits of Gi1, Gi3, G0, Gq, Gs (42, 46 kDa), Gz and beta subunits. The same alpha subunits of G proteins were found in BPM, although alpha subunits of Gi1, Gq, Gs (46 kDa) were located predominantly in the BBM, and the alpha subunit of G0 was found preferentially in BPM. Moreover, in BBM and BPM, a purified antisera raised against the alpha subunits of Gi1 and Gs, detected a 105 kDa protein and a 67 kDa protein, respectively. Interestingly, the 67 kDa protein was preferentially located in BBM, and none of these proteins were detectable in membranes prepared from brain (control). The asymmetrical distribution of the alpha subunits of G proteins among the two different placental bipolar membranes might reflect the very specialized function of these syncytiotrophoblast membranes in ions and nutrients transport from the mother to the fetus.


Subject(s)
Cell Membrane/chemistry , GTP-Binding Proteins/analysis , Microvilli/chemistry , Trophoblasts/ultrastructure , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunoblotting , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...