Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-35300074

RESUMEN

Background: Basic fibroblast growth factor (bFGF)-mediated vascular smooth muscle cell (VSMC) proliferation and migration play an important role in vascular injury-induced neointima formation and subsequent vascular restenosis, a major event that hinders the long-term success of angioplasty. The function of ß3-adrenergic receptors (ß3-ARs) in vascular injury-induced neointima formation has not yet been defined. Objectives: Our current study explored the possible role of ß3-ARs in vascular injury-induced neointima formation by testing its effects on bFGF-induced VSMC migration and proliferation. Methods: ß3-AR expression in rat carotid arteries was examined at 14 days following a balloon catheter-induced injury. The effects of ß3-AR activation on bFGF-induced rat aortic smooth muscle cell proliferation, migration, and signaling transduction (including extracellular-signal-regulated kinase/mitogen activated protein kinase, ERK/MAPK and Protein kinase B, AKT) were tested. Results: We found that vascular injury induced upregulation of ß3-ARs in neointima. Pretreatment of VSMCs with a selective ß3-AR agonist, CL316,243 significantly potentiated bFGF-induced cell migration and proliferation, and ERK and AKT phosphorylation. Our results also revealed that suppressing phosphorylation of ERK and AKT blocked bFGF-induced cell migration and that inhibiting AKT phosphorylation reduced bFGF-mediated cell proliferation. Conclusion: Our results suggest that activation of ß3-ARs potentiates bFGF-mediated effects on VSMCs by enhancing bFGF-mediated ERK and AKT phosphorylation and that ß3-ARs may play a role in vascular injury-induced neointima formation.

2.
Neuroreport ; 31(11): 833-839, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32568774

RESUMEN

Neuroinflammation has been implicated in neurodegenerative diseases and acute brain injuries such as stroke. Monocyte chemotactic protein-1-induced protein-1 (Mcpip1) is a multifunction protein known to have pro-apoptotic or anti-apoptotic actions depending on the nature of experimental settings. However, its role in brain damage after asphyxia in the developing brain has not been studied. We, therefore, explored the role of Mcpip1 in brain injury after hypoxic-ischemia in neonatal mice. At postnatal day 7, Mcpip1-deficient and wild type mice underwent a carotid artery ligation and exposure to hypoxia (8% oxygen). After hypoxic-ischemic insult, we determined the time-course of apoptotic cell death and the expression levels of genes encoding proinflammatory factors. The impact of Mcpip1 on long-term brain damage was assessed 1 week post-hypoxic-ischemia by cresyl violet staining. We found caspase-3 activity was significantly increased in the ipsilateral brain tissues within 12-24 h after hypoxic-ischemia. There was a marked increase in the levels of mRNA transcripts encoding Mcpip1, TNFα, and CCL2 in the ipsilateral brain tissues 6-48 h after hypoxic-ischemia. We found hypoxic-ischemia-induced caspase-3 activity and the levels of the proinflammatory genes were attenuated in Mcpip1-knockout mice compared to wild type mice. Histological assessment revealed that hypoxic-ischemia-induced brain tissue loss was significantly attenuated in the hippocampus of Mcpip1-knockout mice than that of wild type mice (9.0 ± 5.6% vs. 33.9 ± 11.0%, P < 0.05). Our data suggest that Mcpip1 contributes to acute and delayed brain damage, in part, via regulation of neuroinflammation after hypoxic-ischemic insult in the developing mouse brain.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Neuronas/metabolismo , Neuronas/patología , Ribonucleasas/metabolismo , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Artículo en Inglés | MEDLINE | ID: mdl-34909637

RESUMEN

Atherosclerosis involves interactions between inflammation system and dyslipidemia. MCPIP1 (Monocyte Chemotactic Protein induced Protein-1) is induced by proinflammatory molecules and serves as a negative feedback loop in regulating inflammatory responses. Our current study was designed to test the role of MCPIP1 in maintaining lipid homeostasis, the latter a pivotal factor that contributes to the pathogenesis of atherosclerosis. We found that MCPIP1 knockout mice displayed a decrease in levels of serum HDL-cholesterol and total triglycerides but an increase in serum LDL/VLDL-cholesterol levels when compared to wild-type mice. Additionally, ApoA-1 expression was reduced but LPL expression was upregulated in plasma from MCPIP1 knockout mice. The livers from the MCPIP1 knockout mice revealed a decrease in hepatocyte number and an increase in collagen deposition when compared to wild-type mice. These findings suggest that MCPIP1 deficiency can induce liver fibrosis, alter the expression of lipoproteins, and affect transportation and metabolism of lipids, indicating that MCPIP1 is involved in maintaining lipid homeostasis, possibly via negatively regulating inflammatory responses.

4.
J Allergy Clin Immunol ; 142(2): 582-594.e10, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29111212

RESUMEN

BACKGROUND: Asthmatic and allergic inflammation is mediated by TH2 cytokines (IL-4, IL-5, and IL-13). Although we have learned much about how TH2 cells are differentiated, the TH2 checkpoint mechanisms remain elusive. OBJECTIVES: In this study we investigate how monocyte chemotactic protein-induced protein 1 (MCPIP1; encoded by the Zc3h12a gene) regulates IL-5-producing TH2 cell differentiation and TH2-mediated inflammation. METHODS: The functions of Zc3h12a-/- CD4 T cells were evaluated by checking the expression of TH2 cytokines and transcription factors in vivo and in vitro. Allergic airway inflammation of Zc3h12a-/- mice was examined with murine asthma models. In addition, antigen-specific CD4 T cells deficient in MCPIP1 were transferred to wild-type recipient mice, challenged with ovalbumin (OVA) or house dust mite (HDM), and accessed for TH2 inflammation. RESULTS: Zc3h12a-/- mice have spontaneous severe lung inflammation, with an increase in mainly IL-5- and IL-13-producing but not IL-4-producing TH2 cells in the lung. Mechanistically, differentiation of IL-5-producing Zc3h12a-/- TH2 cells is mediated through Notch signaling and Gata3 independent of IL-4. Gata3 mRNA is stabilized in Zc3h12a-/- TH2 cells. MCPIP1 promotes Gata3 mRNA decay through the RNase domain. Furthermore, deletion of MCPIP1 in OVA- or HDM-specific T cells leads to significantly increased TH2-mediated airway inflammation in OVA or HDM murine models of asthma. CONCLUSIONS: Our study reveals that MCPIP1 regulates the development and function of IL-5-producing TH2 cells through the Notch/Gata3 pathway. MCPIP1 represents a new and promising target for the treatment of asthma and other TH2-mediated diseases.


Asunto(s)
Asma/inmunología , Inflamación/inmunología , Hipersensibilidad Respiratoria/inmunología , Ribonucleasas/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factor de Transcripción GATA3/metabolismo , Humanos , Terapia de Inmunosupresión , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Notch/metabolismo , Ribonucleasas/genética , Transducción de Señal , Células Th2/trasplante
5.
Curr Opin Pharmacol ; 33: 47-55, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28527325

RESUMEN

Association of hypercholesterolemia and atherosclerotic cardiovascular disease (ASCVD) is well established. Reducing low-density lipoprotein-cholesterol (LDL-C) and raising high-density lipoprotein-cholesterol (HDL-C) have been the therapeutic targets to reduce the risk of ASCVD. Cholesterol-lowering medications have been used to provide both primary and secondary prevention of ASCVD for many years by reducing the absorption and reabsorption, promoting excretion, or decreasing the synthesis of cholesterol. Within the past five years, several new classes of cholesterol-lowering drugs have been tested and approved for patients with hypercholesterolemia that are not well controlled by conventional therapy (ezetimibe, bile-acid sequestrants, and statins). These drugs include proprotein convertase subtilisin/kexin type 9 (PCSK9) antibodies, apolipoprotein A-100 (Apo B-100) antisense, and microsomal triglyceride transfer protein (MTP) inhibitor. Clinical trials revealed that adding PCSK9 antibodies to the preexisting statin therapy can further reduce LDL-C by 60%. ApoB antisense and MTP inhibitor are currently approved for patients with homozygous familial hypercholesterolemia. Several HDL-raising drugs have also been tested, but the results are not promising. Studies suggest that specifically raising reverse cholesterol transport rather than HDL-C level could be a novel therapeutic approach to reduce cardiovascular risk.


Asunto(s)
Anticolesterolemiantes/farmacología , Anticolesterolemiantes/uso terapéutico , Dislipidemias/tratamiento farmacológico , Animales , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Colesterol/metabolismo , Dislipidemias/complicaciones , Dislipidemias/metabolismo , Humanos , Factores de Riesgo
6.
J Cell Physiol ; 232(1): 122-8, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27035356

RESUMEN

Monocyte chemoattractant protein-1 (MCP-1) has been reported to induce the expression of monocyte chemotactic protein-induced protein 1 (MCPIP1), which undergoes ubiquitination degradation. Therefore, we predict that in vascular smooth muscle (VSMCs), MCPIP1 may be induced by MCP-1 and undergo degradation, which can be inhibited by the proteasome inhibitor, MG132. Our results showed that treatment of human VSMCs with MCP-1 did not increase the expression of MCPIP1. Treatment with MG132, however, elevated MCPIP1 protein levels through stimulation of the gene transcription, but not through increasing protein stability. MCPIP1 expression induced by MG132 was inhibited by α-amanitin inhibition of gene transcription or cycloheximide inhibition of protein synthesis. Our further studies showed that MCPIP1 expression induced by MG132 was inhibited by the inhibitors of AKT and p38 kinase, suggesting a role of the AKT-p38 pathway in MG132 effects. We also found that treatment with MG132 induces apoptosis, but overexpression of MCPIP1 inhibited bromodeoxyuridine (BrdU) incorporation of human VSMCs without induction of significant apoptosis. In summary, MCPIP1 expression is induced by MG132 likely through activation of the AKT-p38 pathway. MCPIP1 inhibits SMC proliferation without induction of apoptosis. J. Cell. Physiol. 232: 122-128, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Leupeptinas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Células Cultivadas , Humanos , Músculo Liso Vascular/citología , Factores de Transcripción/metabolismo
7.
Mol Endocrinol ; 27(11): 1969-79, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24030252

RESUMEN

GH is a potent anabolic and metabolic factor that binds its cell surface receptor (GHR), activating the GHR-associated tyrosine kinase, Janus kinase 2, which phosphorylates and activates the latent transcription factor, signal transducer and activator of transcription 5 (STAT5). Some GH actions are mediated by the elaboration of IGF-1, which exerts effects by binding and activating the heterotetrameric tyrosine kinase growth factor receptor, IGF-1R. In addition to this GH-GHR-IGF-1-IGF-1R scheme, we have demonstrated in primary osteoblasts and in islet ß-cells that then deletion or silencing of IGF-1R results in diminished GH-induced STAT5 phosphorylation, suggesting that the presence of IGF-1R may facilitate GH signaling. In this study, we explore potential roles for protein tyrosine phosphatase activity in modulating GH-induced signaling, comparing conditions in which IGF-1R is present or diminished. We confirm that in mouse primary osteoblasts harboring loxP sites flanking the IGF-1R gene, infection with an adenovirus that expresses the Cre recombinase results in IGF-1R deletion and diminished acute GH-induced STAT5 phosphorylation. Furthermore, we present a new model of IGF-1R silencing, in which expression of short hairpin RNA directed at IGF-1R greatly reduces IGF-1R abundance in LNCaP human prostate cancer cells. In both models, treatment with a chemical inhibitor of protein tyrosine phosphatase-1B (PTP-1B), but not one of src homology region 2 domain-containing phosphotase-1 (SHP-1) and SHP-2, reverses the loss of GH-induced STAT5 phosphorylation in cells lacking IGF-1R but has no effect in cells with intact IGF-1R. Furthermore, expression of either a dominant-negative PTP-1B or the PTP-1B-interacting inhibitory protein, constitutive photomorphogenesis 1, also rescues acute GH-induced STAT5 signaling in IGF-1R-deficient cells but has no effect in IGF-1R replete cells. By expressing a substrate-trapping mutant PTP-1B, we demonstrate that tyrosine phosphorylated Janus kinase-2 is a PTP-1B substrate only in cells lacking IGF-1R. Collectively, our data suggest that IGF-1R positively regulates acute GH signaling by preventing access of PTP-1B activity to Janus kinase 2 and thereby preventing PTP-1B-mediated suppression of GH-induced STAT5 activation.


Asunto(s)
Hormona del Crecimiento/fisiología , Procesamiento Proteico-Postraduccional , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor IGF Tipo 1/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Benzofuranos/farmacología , Células Cultivadas , Humanos , Janus Quinasa 2/metabolismo , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Quinolinas/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
8.
J Caffeine Res ; 3(4): 163-168, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24761282

RESUMEN

Background: Inflammation is considered to be a major initiator to angioplasty-induced vascular restenosis. Proinflammatory cytokines stimulate vascular smooth muscle cell (VSMC) migration and proliferation leading to neointimal hyperplasia. It has been reported that chronic caffeine use suppresses the production of proinflammatory cytokine TNF-α (tumor necrosis factor Alpha) and alters adenosine receptor expression in human neutrophils, indicating that caffeine may attenuate vascular injury-induced inflammation and subsequent neointimal hyperplasia. Our current study was designed to test the hypothesis that chronic caffeine treatment decreases vascular injury-induced neointimal hyperplasia by suppressing VSMC migration and proliferation. Methods and Results: The experiments were carried out using both in vivo (rat carotid artery injury model) and in vitro (VSMCs isolated from rat aorta) models. Male Sprague-Dawley rats that received chronic caffeine treatment (10 and 20 mg/kg per day, through oral gavage) showed a significant decrease in neointimal hyperplasia when compared to rats that received vehicle. To understand the underlying mechanisms, we tested if caffeine inhibits fetal bovine serum (FBS)-induced VSMC migration and proliferation. We found that caffeine substantially suppressed FBS-induced VSMC migration and proliferation. The attenuation of FBS-stimulated cell migration is dose dependent. Conclusion: Together, our results suggest that chronic treatment with high concentrations of caffeine attenuates vascular injury-induced neointimal hyperplasia by suppressing smooth muscle cell migration and proliferation in rats.

9.
J Biol Chem ; 286(48): 41692-41700, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21971051

RESUMEN

It is unclear how stress granule (SG) formation and cellular apoptosis are coordinately regulated. MCPIP1 (monocyte chemotactic protein-induced protein 1), also known as Zc3h12a, is a critical regulator of the inflammatory response and immune homeostasis. However, the role of MCPIP1 in stress response remains unknown. Here, we report that overexpression of MCPIP1 inhibited the assembly of SGs in response to various stresses. Conversely, MCPIP1-deficient splenocytes developed more SGs even without stress. On the other hand, overexpression of MCPIP1 sensitized RAW 264.7 cells to apoptosis under stress, whereas MCPIP1-deficient cells were resistant to stress-induced apoptosis. Mutagenesis study showed that the ability of MCPIP1 to repress SG formation is dependent on its deubiquitinating activity. Consistently, MCPIP1 negatively regulated stress-induced phosphorylation of eIF2α and thus released stress-induced inhibition of protein translation. However, MCPIP1 also inhibited 15-deoxy-Δ(12,14)-prostaglandin J(2)-induced SG formation, which was reported to be independent of eIF2α phosphorylation. Taken together, these results suggest that MCPIP1 coordinates SG formation and apoptosis during cellular stress and may play a critical role in immune homeostasis and resolution of macrophage inflammation.


Asunto(s)
Apoptosis/fisiología , Homeostasis/fisiología , Macrófagos/metabolismo , Ribonucleasas/metabolismo , Estrés Fisiológico/fisiología , Factores de Transcripción/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Mutagénesis , Ribonucleasas/genética , Ribonucleasas/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
10.
Am J Physiol Heart Circ Physiol ; 296(1): H132-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19011046

RESUMEN

We tested the hypothesis that hyperinsulinemia induces the suppression of protein tyrosine phosphatase 1B (PTP1B) function, leading to enhanced PDGF receptor (PDGFR) signaling and neointimal hyperplasia. Rats were implanted with insulin-releasing pellets or sham pellets. Blood glucose levels, insulin levels, food and water intake, body weights, and blood pressures were measured. Neointimal hyperplasia was assessed by computerized morphometry 14 days after carotid balloon injury. PTP1B protein expression in injured arteries was determined via Western blot analysis, whereas PTP1B activity was determined via an immunophosphatase assay. Serum insulin levels were two- to threefold greater in hyperinsulinemic rats, whereas systolic blood pressures, food and water intake, serum triglyceride levels, plasma cortisol levels, and urinary catecholamine levels were not affected. Fourteen days after injury, neointima-to-media area ratios were 0.89 +/- 0.23 and 1.35 +/- 0.22 in control and hyperinsulinemic rats, respectively (P < 0.01). PTP1B protein levels and total PTP1B activity in injured carotid arteries from the insulin-treated group were significantly decreased 7 or 14 days after injury, whereas PTP1B specific activity was decreased only 14 days after injury. These findings were associated with decreased PTP1B mRNA levels and increased PDGFR tyrosyl phosphorylation in insulin-treated rats. These observations support the hypothesis that hyperinsulinemia induces the suppression of PTP1B function, leading to enhanced PDGFR signaling and neointimal hyperplasia.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Hipoglucemiantes/farmacología , Insulina/farmacología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Glucemia/metabolismo , Catecolaminas/sangre , Cateterismo , Hidrocortisona/sangre , Hiperinsulinismo/patología , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Hipertrigliceridemia/sangre , Hipertrigliceridemia/inducido químicamente , Hipoglucemiantes/sangre , Infusiones Intravenosas , Insulina/sangre , Masculino , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
11.
Am J Physiol Heart Circ Physiol ; 295(1): H163-73, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18456732

RESUMEN

Hyperinsulinemia plays a major role in the pathogenesis of vascular disease. Restenosis occurs at an accelerated rate in hyperinsulinemia and is dependent on increased vascular smooth muscle cell movement from media to neointima. PDGF plays a critical role in mediating neointima formation in models of vascular injury. We have reported that PDGF increases the levels of protein tyrosine phosphatase PTP1B and that PTP1B suppresses PDGF-induced motility in cultured cells and that it attenuates neointima formation in injured carotid arteries. Others have reported that insulin enhances the mitogenic and motogenic effects of PDGF in cultured smooth muscle cells and that hyperinsulinemia promotes vascular remodeling. In the present study, we tested the hypothesis that insulin amplifies PDGF-induced cell motility by suppressing the expression and function of PTP1B. We found that chronic but not acute treatment of cells with insulin enhances PDGF-induced motility in differentiated cultured primary rat aortic smooth muscle cells and that it suppresses PDGF-induced upregulation of PTP1B protein. Moreover, insulin suppresses PDGF-induced upregulation of PTP1B mRNA levels, PTP1B enzyme activity, and binding of PTP1B to the PDGF receptor-beta, and it enhances PDGF-induced PDGF receptor phosphotyrosylation. Treatment with insulin induces time-dependent upregulation of phosphatidylinositol 3-kinase (PI3-kinase)-delta and activation of Akt, an enzyme downstream of PI3-kinase. Finally, inhibition of PI3-kinase activity, or its function, by pharmacological or genetic means rescues PTP1B activity in insulin-treated cells. These observations uncover novel mechanisms that explain how insulin amplifies the motogenic capacity of the pivotal growth factor PDGF.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Hiperinsulinismo/enzimología , Insulina/metabolismo , Músculo Liso Vascular/enzimología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Animales , Aorta Torácica/enzimología , Aorta Torácica/patología , Becaplermina , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Humanos , Hiperinsulinismo/patología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 26(3): 501-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16373608

RESUMEN

OBJECTIVE: We have previously reported that vascular injury or treatment of cultured vascular smooth muscle cells with platelet-derived growth factor-BB (PDGF-BB) or fibroblast growth factor-2 (FGF2) increases the levels of protein tyrosine phosphatase (PTP)1B. The current study was designed to test the hypothesis that PTP1B attenuates PDGF- or FGF-induced motility and proliferation of cultured cells, as well as neointima formation in injured rat carotid arteries. METHODS AND RESULTS: Treatment of cultured cells with adenovirus expressing PTP1B decreased PDGF-BB- or FGF2-induced cell motility and blocked PDGF-BB- or FGF2-induced proliferation, whereas expression of dominant negative PTP1B (C215S-PTP1B) uncovered the motogenic effect of subthreshold levels of PDGF-BB or FGF2, increased neointimal and medial cell proliferation, and induced neointimal enlargement after balloon injury. The inhibitory effect of PTP1B directed against PDGF in cultured cells was associated with dephosphorylation of the PDGFbeta receptor. CONCLUSIONS: PTP1B suppresses cell proliferation and motility in cultured smooth muscle cells treated with PDGF-BB or FGF2, and the phosphatase plays a counter-regulatory role in vascular injury-induced cell proliferation and neointima formation. Taken together with previous studies indicating increased PTP1B levels in cells treated with growth factors, the current findings are the first to report the existence of an inhibitory feedback loop involving PDGF or FGF, and PTP1B in blood vessels.


Asunto(s)
Anticoagulantes/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Angioplastia de Balón/efectos adversos , Animales , Aorta Torácica/citología , Apoptosis/efectos de los fármacos , Becaplermina , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , División Celular/efectos de los fármacos , División Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Retroalimentación Fisiológica/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación Enzimológica de la Expresión Génica , Músculo Liso Vascular/efectos de los fármacos , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Túnica Íntima/citología
13.
Am J Physiol Cell Physiol ; 290(4): C1263-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16354758

RESUMEN

Recent data support the hypothesis that reactive oxygen species (ROS) play a central role in the initiation and progression of vascular diseases. An important vasoprotective function related to the regulation of ROS levels appears to be the antioxidant capacity of nitric oxide (NO). We previously reported that treatment with NO decreases phosphotyrosine levels of adapter protein p130(cas) by increasing protein tyrosine phosphatase-proline, glutamate, serine, and threonine sequence protein (PTP-PEST) activity, which leads to the suppression of agonist-induced H(2)O(2) elevation and motility in cultured rat aortic smooth muscle cells (SMCs). The present study was performed to investigate the hypotheses that 1) IGF-I increases the activity of the small GTPase Rac1 as well as H(2)O(2) levels and 2) NO suppresses IGF-I-induced H(2)O(2) elevation by decreasing Rac1 activity via increased PTP-PEST activity and dephosphorylation of p130(cas). We report that IGF-I induces phosphorylation of p130(cas) and activation of Rac1 and that NO attenuates these effects. The effects of NO are mimicked by the overexpression of PTP-PEST or dominant-negative (dn)-p130(cas) and antagonized by the expression of dn-PTP-PEST or p130(cas). We conclude that IGF-I induces rat aortic SMC motility by increasing phosphotyrosine levels of p130(cas) and activating Rac1 and that NO decreases motility by activating PTP-PEST, inducing dephosphorylating p130(cas), and decreasing Rac1 activity. Decreased Rac1 activity lowers intracellular H(2)O(2) levels, thus attenuating cell motility.


Asunto(s)
Proteína Sustrato Asociada a CrK/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Óxido Nítrico/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Aorta/citología , Movimiento Celular , Activación Enzimática , Femenino , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Donantes de Óxido Nítrico/metabolismo , Oxidantes/metabolismo , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12 , Ratas , Ratas Sprague-Dawley , Triazenos/metabolismo
14.
Regul Pept ; 129(1-3): 139-46, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15927709

RESUMEN

This study was done to determine if pituitary adenylate cyclase-activating peptide (PACAP)-immunoreactive nerve fibers occur in cardiac muscle as well as intracardiac ganglia of rats and guinea pigs and to clarify the chronotropic actions of PACAP27 in the same species using isolated heart preparations. PACAP nerve fibers were not detected in atrial or ventricular muscle of rat or guinea pig but a few stained nerve fibers occurred in the atrioventricular bundle of the guinea pig. Stained nerve fibers were prominent in intracardiac ganglia of both species. PACAP27 caused a dose-dependent tachycardia in isolated rat hearts (+39 +/- 3 beats/min with 1 nmol, n = 6). Positive and/or negative chronotropic responses were evoked by PACAP27 in guinea pig heart, depending on dose and prior exposure to the peptide. PACAP27 also caused arrhythmias in several guinea pig hearts. Treatment with atropine eliminated or prevented PACAP-evoked bradycardia and arrhythmias, implicating cholinergic neurons in these responses. Positive chronotropic responses to PACAP were unaffected by beta-adrenergic receptor blockade in either species, suggesting that tachycardia resulted from a direct action on the heart. These observations support the conclusion that endogenous PACAP could have a role in regulating parasympathetic input to the heart but through different mechanisms in rats versus guinea pigs. A direct positive chronotropic influence of endogenous PACAP is unlikely since atrial muscle lacks PACAP-immunoreactive nerve fibers.


Asunto(s)
Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Fibras Nerviosas/metabolismo , Factores de Crecimiento Nervioso/biosíntesis , Neuropéptidos/biosíntesis , Neurotransmisores/biosíntesis , Animales , Relación Dosis-Respuesta a Droga , Femenino , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/inervación , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/inervación , Inmunoquímica , Masculino , Factores de Crecimiento Nervioso/farmacología , Neuropéptidos/farmacología , Neurotransmisores/farmacología , Técnicas de Cultivo de Órganos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Ratas , Especificidad de la Especie , Taquicardia/inducido químicamente , Taquicardia/metabolismo , Taquicardia/patología
15.
Am J Physiol Heart Circ Physiol ; 287(5): H2201-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15271661

RESUMEN

Migration and proliferation of vascular smooth muscle cells are key events in injury-induced neointima formation. Several growth factors and ANG II are thought to be involved in neointima formation. A recent report indicated that vascular injury is associated with increased mRNA levels of protein tyrosine phosphatase (PTP)-1B (PTP-1B). In the present study, we tested the following hypotheses: 1) rat carotid artery injury induces the expression of PTP-1B, Src homology-2 domain phosphatase (SHP-2), and PTP-proline, glutamate, serine, and threonine sequence (PEST) protein; and 2) polypeptide growth factors as well as ANG II increase the levels of tyrosine phosphatases in cultured rat aortic smooth muscle cells. We found that vascular injury induced by balloon catheter increases the protein levels of aforementioned phosphatases and that these effects occur in a PTP specific, as well as temporally and regionally specific, manner. Moreover, treatment of cultured primary rat aortic smooth muscle cells with PDGF or bFGF, but not with IGF1, EGF, or ANG II, increases PTP-1B, SHP-2, and PTP-PEST protein levels. These results suggest that increased PDGF and bFGF levels, occurring after vascular injury, may induce expression of several PTPs.


Asunto(s)
Aorta/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Actinas/metabolismo , Animales , Aorta/efectos de los fármacos , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/enzimología , Células Cultivadas , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
16.
Circ Res ; 91(5): 390-7, 2002 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-12215487

RESUMEN

We have previously reported that SHP-2 upregulation is necessary for NO-stimulated motility in differentiated rat aortic smooth muscle cells. We now test the hypothesis that upregulation of SHP-2 is necessary and sufficient to stimulate cell motility. Overexpression of SHP-2 via recombinant adenoviral vector stimulated motility to the same extent as NO, whereas the expression of C463S-SHP-2, the dominant-negative SHP-2 allele, blocked the motogenic effect of NO. On the basis of previous studies, we next tested the hypothesis that NO decreases RhoA activity and that this event is necessary and sufficient to explain NO-induced motogenesis. We found that NO decreased RhoA activity in a concentration-dependent manner. Moreover, a dominant-negative SHP-2 allele, DSH2, blocked the NO-induced inhibition of RhoA activity, indicating that upregulation of SHP-2 is necessary for this event. Expression of G14V-RhoA, the constitutively active RhoA allele, decreased cell motility and blocked the motogenic effect of NO, whereas the expression of T19N-RhoA, the dominant-negative RhoA allele, increased cell motility to an extent similar to that induced by NO. Dominant-negative RhoA reversed the effect of dominant-negative SHP-2, indicating that RhoA functions downstream from SHP-2. To investigate events downstream from RhoA, we treated cells with fasudil, a selective Rho kinase inhibitor, and found that it increased cell motility. These results indicate that upregulation of SHP-2, leading to downregulation of RhoA, which is followed by decreased Rho kinase activity, is a sequence of events necessary and sufficient to explain NO-induced cell motility in differentiated aortic smooth muscle cells. The results may be of relevance to in vivo events such as neointimal formation, angiogenesis, and vasculogenesis.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Movimiento Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Penicilamina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Aorta/citología , Aorta/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Vectores Genéticos/genética , Péptidos y Proteínas de Señalización Intracelular , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Mutación , Óxido Nítrico/fisiología , Penicilamina/farmacología , Fragmentos de Péptidos/farmacología , Fosfotirosina/efectos de los fármacos , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-12232609

RESUMEN

The present study reports the structural and functional changes of myocardial muscarinic receptor during early and late septic shock, Septic shock was induced by caecum ligation and puncture (CLP). The results showed that the number of M receptor on sarcolemma (SL) increased and that on the light vesicle (LV) decreased during early septic shock. The (3)H-QNB binding of M receptor on SL increased by 33.37%. By contrast during late septic shock, the number of M receptor on LV increased and that on SL decreased. The (3)H-QNB binding of LV M receptor was increased by 29.26%. At the same time phosphorylation of the M receptor was decreased during early septic shock and increased during late septic shock. These results suggest that the changes of M receptors may be related to the myocardial dysfunction during septic shock.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA