Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biomed Biotechnol ; 2012: 406273, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23193360

RESUMEN

Since its discovery as a src kinase substrate more than three decades ago, appreciation for the physiologic functions of annexin A2 and its associated proteins has increased dramatically. With its binding partner S100A10 (p11), A2 forms a cell surface complex that regulates generation of the primary fibrinolytic protease, plasmin, and is dynamically regulated in settings of hemostasis and thrombosis. In addition, the complex is transcriptionally upregulated in hypoxia and promotes pathologic neoangiogenesis in the tissues such as the retina. Dysregulation of both A2 and p11 has been reported in examples of rodent and human cancer. Intracellularly, A2 plays a critical role in endosomal repair in postarthroplastic osteolysis, and intracellular p11 regulates serotonin receptor activity in psychiatric mood disorders. In human studies, the A2 system contributes to the coagulopathy of acute promyelocytic leukemia, and is a target of high-titer autoantibodies in patients with antiphospholipid syndrome, cerebral thrombosis, and possibly preeclampsia. Polymorphisms in the human ANXA2 gene have been associated with stroke and avascular osteonecrosis of bone, two severe complications of sickle cell disease. Together, these new findings suggest that manipulation of the annexin A2/S100A10 system may offer promising new avenues for treatment of a spectrum of human disorders.


Asunto(s)
Anexina A2/metabolismo , Enfermedad , Salud , Proteínas S100/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos
2.
J Neurosci ; 28(10): 2383-93, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18322085

RESUMEN

Neurogenesis, the division, migration, and differentiation of new neurons, occurs throughout life. Brain derived neurotrophic factor (BDNF) has been identified as a potential signaling molecule regulating neurogenesis in the subventricular zone (SVZ), but its functional consequences in vivo have not been well defined. We report marked and unexpected deficits in survival but not proliferation of newly born cells of adult knock-in mice containing a variant form of BDNF [a valine (Val) to methionine (Met) substitution at position 66 in the prodomain of BDNF (Val66Met)], a genetic mutation shown to lead to a selective impairment in activity-dependent BDNF secretion. Utilizing knock-out mouse lines, we identified BDNF and tyrosine receptor kinase B (TrkB) as the critical molecules for the observed impairments in neurogenesis, with p75 knock-out mice showing no effect on cell proliferation or survival. We then localized the activated form of TrkB to a discrete population of cells, type A migrating neuroblasts, and demonstrate a decrease in TrkB phosphorylation in the SVZ of Val66Met mutant mice. With these findings, we identify TrkB signaling, potentially through activity dependent release of BDNF, as a critical step in the survival of migrating neuroblasts. Utilizing a behavioral task shown to be sensitive to disruptions in olfactory bulb neurogenesis, we identified specific impairments in spontaneous olfactory discrimination, but not general olfactory sensitivity or habituation to olfactory stimuli in BDNF mutant mice. Through these observations, we have identified novel links between genetic variant BDNF and adult neurogenesis in vivo, which may contribute to significant impairments in olfactory function.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Aprendizaje Discriminativo/fisiología , Variación Genética , Bulbo Olfatorio/citología , Bulbo Olfatorio/crecimiento & desarrollo , Vías Olfatorias/fisiología , Sustitución de Aminoácidos/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Movimiento Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Células Cultivadas , Masculino , Metionina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bulbo Olfatorio/fisiología , Estructura Terciaria de Proteína/genética , Receptor trkB/genética , Células Madre/fisiología , Valina/genética
3.
J Neurosci ; 26(20): 5288-300, 2006 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-16707781

RESUMEN

Studies showing that neurotrophin binding to p75NTR can promote cell survival in the absence of Trk (tropomyosin-related kinase) receptors, together with recent structural data indicating that NGF may bind to p75NTR in a monovalent manner, raise the possibility that small molecule p75NTR ligands that positively regulate survival might be found. A pharmacophore designed to capture selected structural and physical chemical features of a neurotrophin domain known to interact with p75NTR was applied to in silico screening of small molecule libraries. Small, nonpeptide, monomeric compounds were identified that interact with p75NTR. In cells showing trophic responses to neurotrophins, the compounds promoted survival signaling through p75NTR-dependent mechanisms. In cells susceptible to proneurotrophin-induced death, compounds did not induce apoptosis but inhibited proneurotrophin-mediated death. These studies identify a unique range of p75NTR behaviors that can result from isolated receptor liganding and establish several novel therapeutic leads.


Asunto(s)
Apoptosis/efectos de los fármacos , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Precursores de Proteínas/antagonistas & inhibidores , Receptor de Factor de Crecimiento Nervioso/agonistas , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Evaluación Preclínica de Medicamentos/métodos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Isoleucina/análogos & derivados , Isoleucina/farmacología , Ligandos , Ratones , Estructura Molecular , Peso Molecular , Morfolinas/farmacología , Células 3T3 NIH , Factor de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/síntesis química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/síntesis química , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Células PC12 , Precursores de Proteínas/metabolismo , Estructura Terciaria de Proteína/fisiología , Ratas , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
4.
J Neurosci ; 25(22): 5455-63, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930396

RESUMEN

Brain-derived neurotrophic factor (BDNF) is best characterized for critical roles in neuronal survival, differentiation, and synaptic modulation mediated by the TrkB receptor tyrosine kinase. Developmentally regulated death signaling by BDNF has also been demonstrated via activation of p75NTR. Because recent studies suggest that proNGF, the precursor form of NGF, is more active than mature NGF in inducing apoptosis after binding to p75NTR and a coreceptor, sortilin, we asked whether the precursor of BDNF (proBDNF) is also a proapoptotic ligand in the nervous system. proBDNF is secreted by cultured neurons, and recombinant proBDNF binds to sortilin. In sympathetic neurons coexpressing sortilin and p75NTR, we found that proBDNF is an apoptotic ligand that induces death at subnanomolar concentrations. In contrast, mature BDNF, but not proBDNF, is effective in inducing TrkB phosphorylation. proBDNF effects are dependent on cellular coexpression of both p75NTR and sortilin, because neurons deficient in p75NTR are resistant to proBDNF-induced apoptosis, and competitive antagonists of sortilin block sympathetic neuron death. Moreover, addition of preformed complexes of soluble sortilin and proBDNF failed to induce apoptosis of cells coexpressing both sortilin and p75NTR, suggesting that interaction of proBDNF with both receptors on the cell surface is required to initiate cell death. Together with our past findings, these data suggest that the neurotrophin family is capable of modulating diverse biological processes via differential processing of the proneurotrophins.


Asunto(s)
Apoptosis , Factor Neurotrófico Derivado del Encéfalo/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Precursores de Proteínas/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Animales , Encéfalo/citología , Línea Celular , Humanos , Ligandos , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkB/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factor de Crecimiento Nervioso , Células de Schwann/citología , Células de Schwann/metabolismo , Ganglio Cervical Superior/citología
5.
Circulation ; 112(23): 3644-53, 2005 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-16330706

RESUMEN

BACKGROUND: Accumulation of macrophages and smooth muscle cells in the vascular wall is critical for the development of atherosclerotic lesions. Although much is known about the factors that regulate macrophage recruitment to the vascular wall, the ability of growth factors to regulate smooth muscle cell recruitment in lesion development in vivo is unclear. Our previous studies demonstrated that neurotrophins and their receptors, the Trk receptor tyrosine kinases, are potent chemotactic factors for smooth muscle cells, and the expression of brain-derived neurotrophic factor (BDNF) and its cognate receptor, TrkB, is upregulated in human atherosclerotic lesions. METHODS AND RESULTS: TrkB(+/-) mice on a 129/B6 background were backcrossed to apolipoprotein E (ApoE)-null (ApoE(-/-)) mice on the C57Bl/6 background for 6 to 8 generations. Immunohistochemical analysis demonstrated BDNF immunoreactivity in areas of macrophage and smooth muscle cell infiltration, whereas TrkB immunoreactivity was predominant in areas of neointimal smooth muscle cells. Moreover, haplodeficient expression of TrkB in ApoE(-/-) mice was associated with a 30% to 40% reduction in lesion size compared with ApoE(-/-) mice with normal expression of TrkB and a 45% decrease in smooth muscle cell accumulation in the lesions. Finally, reconstitution with bone marrow from ApoE(-/-) mice with normal TrkB expression did not restore lesion development in TrKB(+/-)/ApoE(-/-) mice. CONCLUSIONS: These results suggest that TrkB expression on smooth muscle cells contributes to lesion development in the cholesterol-fed ApoE-null mutant mouse. These data demonstrate, for the first time, a role for the neurotrophin TrkB receptor in atherosclerotic lesion development.


Asunto(s)
Arteriosclerosis/etiología , Receptor trkB/genética , Receptor trkB/fisiología , Animales , Apolipoproteínas E/deficiencia , Arteriosclerosis/patología , Factor Neurotrófico Derivado del Encéfalo/análisis , Quimiotaxis , Colesterol/administración & dosificación , Regulación de la Expresión Génica/fisiología , Macrófagos/química , Macrófagos/fisiología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/fisiología
6.
Circ Res ; 91(6): 494-500, 2002 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-12242267

RESUMEN

Apoptosis of neointimal smooth muscle cells is a well-recognized component of the pathogenesis of vascular lesions. In recent studies, we have identified the neurotrophin receptor, p75(NTR), as a mediator of apoptosis of neointimal smooth muscle cells. Neurotrophin ligands and p75(NTR) are selectively expressed in areas of atherosclerotic lesions with increased smooth muscle cell apoptosis and the neurotrophins are potent apoptotic agents for p75(NTR)-expressing smooth muscle cells in vitro. In the present study, we directly assess the role of p75(NTR) in lesion development in the flow-restricted carotid artery, a model of murine vascular injury. Ligation of the left carotid artery resulted in a 3- to 4-fold increase in lesion development in p75(NTR)-null mutant mice as compared with wild-type mice. The increase in lesion size was associated with a 70% decrease in apoptosis of neointimal smooth muscle cells, as assessed by in situ TUNEL analysis. These data suggest that under conditions of flow restriction, p75(NTR) activation impairs lesion formation by promoting smooth muscle cell apoptosis. These results further implicate p75(NTR) as an important regulator of smooth muscle cell apoptosis and lesion development after vascular injury.


Asunto(s)
Apoptosis , Arterias Carótidas/patología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Arterias Carótidas/metabolismo , Arterias Carótidas/fisiopatología , Femenino , Genotipo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Receptor de Factor de Crecimiento Nervioso , Receptores de Factor de Crecimiento Nervioso/genética , Factores de Tiempo , Túnica Íntima/metabolismo , Túnica Íntima/patología , Túnica Íntima/fisiopatología
7.
Front Biosci ; 8: s1181-6, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12957860

RESUMEN

Angiogenesis is a highly coordinated physiological process in which new blood vessels are formed to meet the oxygenation demands of local tissues. Several classes of growth factors, including members of the vascular endothelial growth factor family, angiopoietins, platelet derived growth factor, fibroblast growth factors and ephrins have been implicated in regulating specific aspects of angiogenesis, both during embryonic development and in response to injury. This review focuses on a distinct family of growth factors, the neurotrophins, and their receptors as newly identified angiogenic molecules. The expression of neurotrophins and their receptors are regulated both temporally and spatially by the vasculature of the embryo and adult, and dynamic changes occur following vascular injury. Recent studies that identify the vascular cells responsive to neurotrophins, that genetically dissect neurotrophin actions in vessel development and remodeling, and that uncover neurotrophin effects in models of tissue ischemia are discussed.


Asunto(s)
Neovascularización Fisiológica/fisiología , Factores de Crecimiento Nervioso/fisiología , Animales , Humanos
8.
Am J Pathol ; 168(1): 349-62, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16400036

RESUMEN

Nitric oxide (NO) modulates the biological levels of arachidonate-derived cell signaling molecules by either enhancing or suppressing the activity of prostaglandin H(2) isoforms (PGHS-1 and PGHS-2). Whether NO activates or suppresses PGHS activity is determined by alternative protein modifications mediated by NO and NO-derived species. Here, we show that inducible NO synthase (iNOS) and PGHS-1 co-localize in atherosclerotic lesions of ApoE(-/-) mouse aortae. Immunoblotting and immunohistochemistry revealed Tyr nitration in PGHS-1 in aortic lesions but markedly less in adjacent nonlesion tissue. PGHS-2 was also found in lesions, but 3-nitrotyrosine incorporation was not detected. 3-Nitrotyrosine formation in proteins is considered a hallmark reaction of peroxynitrite, which can form via NO-superoxide reactions in an inflammatory setting. That iNOS-derived NO is essential for 3-nitrotyrosine modification of PGHS-1 was confirmed by the absence of 3-nitrotyrosine in lesions from ApoE(-/-)iNOS(-/-) mice. Mass spectrometric studies specifically identified the active site residue Tyr385 as a 3-nitrotyrosine modification site in purified PGHS-1 exposed to peroxynitrite. PGHS-mediated eicosanoid (PGE(2)) synthesis was more than fivefold accelerated in cultured iNOS(-/-) versus iNOS-expressing mouse aortic smooth muscle cells, suggesting that iNOS-derived NO markedly suppresses PGHS activity in vascular cells. These results further suggest a regulatory role of iNOS in eicosanoid biosynthesis in human atherosclerotic lesions.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Eicosanoides/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Northern Blotting , Western Blotting , Células Cultivadas , Enfermedad de la Arteria Coronaria/patología , Femenino , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Tirosina/análogos & derivados , Tirosina/metabolismo
9.
J Biol Chem ; 280(14): 13801-8, 2005 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15668238

RESUMEN

Activation of the p75 neurotrophin receptor leads to a variety of effects within the nervous system, including neuronal apoptosis. Both c-Jun N-terminal kinase (JNK) and the tumor suppressor p53 have been reported to be critical for this receptor to induce cell death; however, the mechanisms by which p75 activates these pathways is undetermined. Here we report that the neurotrophin receptor interacting factor (NRIF) is necessary for p75-dependent JNK activation and apoptosis. Upon nerve growth factor withdrawal, nrif-/- sympathetic neurons underwent apoptosis, whereas p75-mediated death was completely abrogated. The lack of cell death correlated with a lack of JNK activation in the nrif-/- neurons, suggesting that NRIF is a selective mediator for p75-dependent JNK activation and apoptosis. Moreover, we document that NRIF expression is sufficient to induce cell death through a mechanism that requires p53. Taken together, these results establish NRIF as an essential component of the p75 apoptotic pathway.


Asunto(s)
Apoptosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/fisiología , Fibras Adrenérgicas/metabolismo , Animales , Caspasas/metabolismo , Células Cultivadas , Proteínas de Unión al ADN , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Ratas , Receptor de Factor de Crecimiento Nervioso , Receptores de Factor de Crecimiento Nervioso/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
J Biol Chem ; 277(3): 2353-9, 2002 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-11698409

RESUMEN

In response to vascular injury, smooth muscle cells migrate from the media into the intima, where they contribute to the development of neointimal lesions. Increased matrix metalloproteinase (MMP) expression contributes to the migratory response of smooth muscle cells by releasing them from their surrounding extracellular matrix. MMPs may also participate in the remodeling of extracellular matrix in vascular lesions that could lead to plaque weakening and subsequent rupture. Neurotrophins and their receptors, the Trk family of receptor tyrosine kinases, are expressed in neointimal lesions, where they induce smooth muscle cell migration. We now report that nerve growth factor (NGF)-induced activation of the TrkA receptor tyrosine kinase induces MMP-9 expression in both primary cultured rat aortic smooth muscle cells and in a smooth muscle cell line genetically manipulated to express TrkA. The response to NGF was specific for MMP-9 expression, as the expression of MMP-2, MMP-3, or the tissue inhibitor of metalloproteinase-2 was not changed. Activation of the Shc/mitogen-activated protein kinase pathway mediates the induction of MMP-9 in response to NGF, as this response is abrogated in cells expressing a mutant TrkA receptor that does not bind Shc and by pretreatment of cells with the MEK-1 inhibitor, U0126. Thus, these results indicate that the neurotrophin/Trk receptor system, by virtue of its potent chemotactic activity for smooth muscle cells and its ability to induce MMP-9 expression, is a critical mediator in the remodeling that occurs in the vascular wall in response to injury.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Animales , Células Cultivadas , Activación Enzimática , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , ARN Mensajero/genética , Ratas , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA