Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Oncogene ; 39(5): 1049-1062, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31582836

RESUMEN

Semaphorins, specifically type IV, are important regulators of axonal guidance and have been increasingly implicated in poor prognoses in a number of different solid cancers. In conjunction with their cognate PLXNB family receptors, type IV members have been increasingly shown to mediate oncogenic functions necessary for tumor development and malignant spread. In this study, we investigated the role of semaphorin 4C (SEMA4C) in osteosarcoma growth, progression, and metastasis. We investigated the expression and localization of SEMA4C in primary osteosarcoma patient tissues and its tumorigenic functions in these malignancies. We demonstrate that overexpression of SEMA4C promotes properties of cellular transformation, while RNAi knockdown of SEMA4C promotes adhesion and reduces cellular proliferation, colony formation, migration, wound healing, tumor growth, and lung metastasis. These phenotypic changes were accompanied by reductions in activated AKT signaling, G1 cell cycle delay, and decreases in expression of mesenchymal marker genes SNAI1, SNAI2, and TWIST1. Lastly, monoclonal antibody blockade of SEMA4C in vitro mirrored that of the genetic studies. Together, our results indicate a multi-dimensional oncogenic role for SEMA4C in metastatic osteosarcoma and more importantly that SEMA4C has actionable clinical potential.


Asunto(s)
Neoplasias Óseas/patología , Progresión de la Enfermedad , Osteosarcoma/patología , Semaforinas/metabolismo , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/secundario , Metástasis de la Neoplasia , Semaforinas/deficiencia , Semaforinas/genética
2.
J Immunol ; 204(1): 128-136, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31776203

RESUMEN

Semaphorin 3E (Sema3E) is a secreted protein that was initially discovered as a neuronal guidance cue. Recent evidence showed that Sema3E plays an essential role in regulating the activities of various immune cells. However, the exact role of Sema3E in macrophage function, particularly during inflammation, is not fully understood. We studied the impact of Sema3E gene deletion on macrophage function during the LPS-induced acute inflammatory response. We found that Sema3E-deficient (Sema3e-/- ) mice were better protected from LPS-induced acute inflammation as exemplified by their superior clinical score and effective temperature control compared with their wild-type littermates. This superior control of inflammatory response in Sema3e-/- mice was associated with significantly lower phosphorylation of ERK1/2, AKT, STAT3, and NF-κB, and a concomitant reduction in inducible NO synthase expression and production of TNF and IL-6 compared with their Sema3e+/+ littermates. Sema3e-/- mice also contained significantly higher numbers of activated macrophages compared with their Sema3e+/+ littermates at both baselines and after LPS challenge. In vivo-specific deletion of the Sema3E high-affinity receptor, plexinD1, on macrophages led to the improvement in clinical disease following exposure to a lethal dose of LPS. Collectively, our data show that Sema3E plays an essential role in dampening the early inflammatory response to LPS by regulating macrophage function, suggesting an essential role of this pathway in macrophage inflammatory response.


Asunto(s)
Inflamación/inmunología , Macrófagos/inmunología , Semaforinas/inmunología , Animales , Células Cultivadas , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Noqueados , Ratones Transgénicos , Semaforinas/deficiencia
3.
J Biol Chem ; 294(21): 8336-8347, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30979723

RESUMEN

Primary hyperparathyroidism (PHPT) is a common endocrinopathy characterized by hypercalcemia and elevated levels of parathyroid hormone. The primary cause of PHPT is a benign overgrowth of parathyroid tissue causing excessive secretion of parathyroid hormone. However, the molecular etiology of PHPT is incompletely defined. Here, we demonstrate that semaphorin3d (Sema3d), a secreted glycoprotein, is expressed in the developing parathyroid gland in mice. We also observed that genetic deletion of Sema3d leads to parathyroid hyperplasia, causing PHPT. In vivo and in vitro experiments using histology, immunohistochemistry, biochemical, RT-qPCR, and immunoblotting assays revealed that Sema3d inhibits parathyroid cell proliferation by decreasing the epidermal growth factor receptor (EGFR)/Erb-B2 receptor tyrosine kinase (ERBB) signaling pathway. We further demonstrate that EGFR signaling is elevated in Sema3d-/- parathyroid glands and that pharmacological inhibition of EGFR signaling can partially rescue the parathyroid hyperplasia phenotype. We propose that because Sema3d is a secreted protein, it may be possible to use recombinant Sema3d or derived peptides to inhibit parathyroid cell proliferation causing hyperplasia and hyperparathyroidism. Collectively, these findings identify Sema3d as a negative regulator of parathyroid growth.


Asunto(s)
Proliferación Celular , Hiperparatiroidismo Primario/epidemiología , Glándulas Paratiroides/embriología , Semaforinas/deficiencia , Transducción de Señal , Animales , Receptores ErbB/genética , Receptores ErbB/metabolismo , Hiperparatiroidismo Primario/genética , Hiperparatiroidismo Primario/patología , Ratones , Ratones Noqueados , Glándulas Paratiroides/patología , Semaforinas/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 38(2): 335-343, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29269512

RESUMEN

OBJECTIVE: Accumulating evidence suggests a role of semaphorins in vascular homeostasis. Here, we investigate the role of Sema7A (semaphorin 7A) in atherosclerosis and its underlying mechanism. APPROACH AND RESULTS: Using genetically engineered Sema7A-/-ApoE-/- mice, we showed that deletion of Sema7A attenuates atherosclerotic plaque formation primarily in the aorta of ApoE-/- mice on a high-fat diet. A higher level of Sema7A in the atheroprone lesser curvature suggests a correlation of Sema7A with disturbed flow. This notion is supported by elevated Sema7A expression in human umbilical venous endothelial cells either subjected to oscillatory shear stress or treated with the PKA (protein kinase A)/CREB (cAMP response element-binding protein) inhibitor H89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide·2HCl hydrate). Further studies using the partial carotid artery ligation model showed that disturbed flow in the left carotid artery of Sema7A+/+ApoE-/- mice promoted the expression of endothelial Sema7A and cell adhesion molecules, leukocyte adhesion, and plaque formation, whereas such changes were attenuated in Sema7A-/-ApoE-/- mice. Further studies showed that blockage of ß1 integrin, a known Sema7A receptor, or inhibition of FAK (focal adhesion kinase), MEK1/2 (mitogen-activated protein kinase kinase 1/2), or NF-κB (nuclear factor-κB) significantly reduced the expression of cell adhesion molecules and THP-1 (human acute monocytic leukemia cell line) monocyte adhesion in Sema7A-overexpressing human umbilical venous endothelial cells. Studies using chimeric mice suggest that vascular, most likely endothelial, Sema7A plays a major role in atherogenesis. CONCLUSIONS: Our findings indicate a significant role of Sema7A in atherosclerosis by mediating endothelial dysfunction in a ß1 integrin-dependent manner.


Asunto(s)
Antígenos CD/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Células Endoteliales/metabolismo , Integrina beta1/metabolismo , Mecanotransducción Celular , Semaforinas/metabolismo , Animales , Antígenos CD/genética , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Rodamiento de Leucocito , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , FN-kappa B/metabolismo , Placa Aterosclerótica , Flujo Sanguíneo Regional , Semaforinas/deficiencia , Semaforinas/genética , Células THP-1 , Regulación hacia Arriba
5.
J Cell Biol ; 216(1): 199-215, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-28007914

RESUMEN

Semaphorins comprise a large family of ligands that regulate key cellular functions through their receptors, plexins. In this study, we show that the transmembrane semaphorin 4A (Sema4A) can also function as a receptor, rather than a ligand, and transduce signals triggered by the binding of Plexin-B1 through reverse signaling. Functionally, reverse Sema4A signaling regulates the migration of various cancer cells as well as dendritic cells. By combining mass spectrometry analysis with small interfering RNA screening, we identify the polarity protein Scrib as a downstream effector of Sema4A. We further show that binding of Plexin-B1 to Sema4A promotes the interaction of Sema4A with Scrib, thereby removing Scrib from its complex with the Rac/Cdc42 exchange factor ßPIX and decreasing the activity of the small guanosine triphosphatase Rac1 and Cdc42. Our data unravel a role for Plexin-B1 as a ligand and Sema4A as a receptor and characterize a reverse signaling pathway downstream of Sema4A, which controls cell migration.


Asunto(s)
Movimiento Celular , Células Dendríticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Semaforinas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Animales , Genotipo , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Espectrometría de Masas , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Interferencia de ARN , Receptores de Superficie Celular/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Semaforinas/deficiencia , Semaforinas/genética , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo
6.
J Virol ; 89(23): 11834-44, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26378166

RESUMEN

UNLABELLED: Hantaan virus (HTNV) infection can cause a severe lethal hemorrhagic fever with renal syndrome (HFRS) in humans. CD8(+) T cells play a critical role in combating HTNV infections. However, the contributions of different CD8(+) T cell subsets to the immune response against viral infection are poorly understood. Here, we identified a novel subset of CD8(+) T cells characterized by the CD8(low) CD100(-) phenotype in HFRS patients. The CD8(low) CD100(-) subset accounted for a median of 14.3% of the total CD8(+) T cells in early phase of HFRS, and this percentage subsequently declined in the late phase of infection, whereas this subset was absent in healthy controls. Furthermore, the CD8(low) CD100(-) cells were associated with high activation and expressed high levels of cytolytic effector molecules and exhibited a distinct expression profile of effector CD8(+) T cells (CCR7(+/-) CD45RA(-) CD127(high) CD27(int) CD28(low) CD62L(-)). When stimulated with specific HTNV nucleocapsid protein-derived peptide pools, most responding CD8(+) cells (gamma interferon [IFN-γ] positive and/or tumor necrosis factor alpha [TNF-α] positive) were CD8(low) CD100(-) cells. The frequency of CD8(low) CD100(-) cells among HTNV-specific CD8(+) T cells was higher in milder cases than in more severe cases. Importantly, the proportion of the CD8(low) CD100(-) subset among CD8(+) T cells in early phase of HFRS was negatively correlated with the HTNV viral load, suggesting that CD8(low) CD100(-) cells may be associated with viral clearance. The contraction of the CD8(low) CD100(-) subset in late phase of infection may be related to the consistently high expression levels of PD-1. These results may provide new insights into our understanding of CD8(+) T cell-mediated protective immunity as well as immune homeostasis after HTNV infection in humans. IMPORTANCE: CD8(+) T cells play important roles in the antiviral immune response. We found that the proportion of CD8(low) CD100(-) cells among CD8(+) T cells from HFRS patients was negatively correlated with the HTNV viral load, and the frequency of CD8(low) CD100(-) cells among virus-specific CD8(+) T cells was higher in milder HFRS cases than in more severe cases. These results imply a beneficial role for the CD8(low) CD100(-) cell subset in viral control during human HTNV infection.


Asunto(s)
Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica/inmunología , Virus Hantaan/inmunología , Fiebre Hemorrágica con Síndrome Renal/inmunología , Semaforinas/inmunología , Subgrupos de Linfocitos T/inmunología , Anticuerpos Monoclonales , Linfocitos T CD8-positivos/virología , China , Citometría de Flujo , Fluoresceína-5-Isotiocianato , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Semaforinas/deficiencia , Estadísticas no Paramétricas , Subgrupos de Linfocitos T/metabolismo
7.
J Clin Invest ; 125(7): 2661-76, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26053665

RESUMEN

In mammals, the outflow tract (OFT) of the developing heart septates into the base of the pulmonary artery and aorta to guide deoxygenated right ventricular blood into the lungs and oxygenated left ventricular blood into the systemic circulation. Accordingly, defective OFT septation is a life-threatening condition that can occur in both syndromic and nonsyndromic congenital heart disease. Even though studies of genetic mouse models have previously revealed a requirement for VEGF-A, the class 3 semaphorin SEMA3C, and their shared receptor neuropilin 1 (NRP1) in OFT development, the precise mechanism by which these proteins orchestrate OFT septation is not yet understood. Here, we have analyzed a complementary set of ligand-specific and tissue-specific mouse mutants to show that neural crest-derived SEMA3C activates NRP1 in the OFT endothelium. Explant assays combined with gene-expression studies and lineage tracing further demonstrated that this signaling pathway promotes an endothelial-to-mesenchymal transition that supplies cells to the endocardial cushions and repositions cardiac neural crest cells (NCCs) within the OFT, 2 processes that are essential for septal bridge formation. These findings elucidate a mechanism by which NCCs cooperate with endothelial cells in the developing OFT to enable the postnatal separation of the pulmonary and systemic circulation.


Asunto(s)
Tabiques Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Cresta Neural/metabolismo , Neuropilina-1/metabolismo , Semaforinas/metabolismo , Animales , Apoptosis , Proliferación Celular , Endotelio Vascular/citología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Femenino , Tabiques Cardíacos/citología , Tabiques Cardíacos/metabolismo , Ventrículos Cardíacos/embriología , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Cresta Neural/embriología , Neuropilina-1/deficiencia , Neuropilina-1/genética , Embarazo , Semaforinas/deficiencia , Semaforinas/genética , Transducción de Señal , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular/deficiencia , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25839327

RESUMEN

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Asunto(s)
Epistasis Genética/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforinas/genética , Animales , Secuencia de Bases , Estudio de Asociación del Genoma Completo , Ratones , Datos de Secuencia Molecular , Semaforinas/deficiencia , Semaforinas/metabolismo , Análisis de Secuencia de ADN
9.
Blood ; 120(19): 4104-15, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-23007403

RESUMEN

Formation of new vessels during development and in the mature mammal generally proceeds through angiogenesis. Although a variety of molecules and signaling pathways are known to underlie endothelial cell sprouting and remodeling during angiogenesis, many aspects of this complex process remain unexplained. Here we show that the transmembrane semaphorin6A (Sema6A) is expressed in endothelial cells, and regulates endothelial cell survival and growth by modulating the expression and signaling of VEGFR2, which is known to maintain endothelial cell viability by autocrine VEGFR signaling. The silencing of Sema6A in primary endothelial cells promotes cell death that is not rescued by exogenous VEGF-A or FGF2, attributable to the loss of prosurvival signaling from endogenous VEGF. Analyses of mouse tissues demonstrate that Sema6A is expressed in angiogenic and remodeling vessels. Mice with null mutations of Sema6A exhibit significant defects in hyaloid vessels complexity associated with increased endothelial cell death, and in retinal vessels development that is abnormally reduced. Adult Sema6A-null mice exhibit reduced tumor, matrigel, and choroidal angiogenesis compared with controls. Sema6A plays important roles in development of the nervous system. Here we show that it also regulates vascular development and adult angiogenesis.


Asunto(s)
Neovascularización Fisiológica/genética , Semaforinas/genética , Semaforinas/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Coroides/irrigación sanguínea , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Noqueados , Retina/metabolismo , Retina/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Semaforinas/deficiencia , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Glia ; 60(10): 1590-604, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22777942

RESUMEN

Myelination is regulated by extracellular proteins, which control interactions between oligodendrocytes and axons. Semaphorins are repulsive axon guidance molecules, which control the migration of oligodendrocyte precursors during normal development and possibly in demyelinating diseases. We show here that the transmembrane semaphorin 6A (Sema6A) is highly expressed by myelinating oligodendrocytes in the postnatal mouse brain. In adult mice, Sema6A expression is upregulated in demyelinating lesions in cuprizone-treated mice. The analysis of the optic nerve and anterior commissure of Sema6A-deficient mice revealed a marked delay of oligodendrocyte differentiation. Accordingly, the development of the nodes of Ranvier is also transiently delayed. We also observed an arrest in the in vitro differentiation of purified oligodendrocytes lacking Sema6A, with a reduction of the expression level of Myelin Basic Protein. Their morphology is also abnormal, with less complex and ramified processes than wild-type oligodendrocytes. In myelinating co-cultures of dorsal root ganglion neurons and purified oligodendrocytes we found that myelination is perturbed in absence of Sema6A. These results suggest that Sema6A might have a role in myelination by controlling oligodendrocyte differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Enfermedades Desmielinizantes/patología , Regulación del Desarrollo de la Expresión Génica/genética , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Semaforinas/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos de Diferenciación/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/citología , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Ganglios Espinales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/toxicidad , Mutación/fisiología , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/efectos de los fármacos , Embarazo , ARN Mensajero/metabolismo , Nódulos de Ranvier/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Superficie Celular/deficiencia , Semaforinas/deficiencia , Células Madre/fisiología , Factores de Tiempo , Factores de Transcripción/metabolismo
11.
Hum Mol Genet ; 20(24): 4759-74, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21903667

RESUMEN

Reproduction in mammals is dependent on the function of specific neurons that secrete gonadotropin-releasing hormone-1 (GnRH-1). These neurons originate prenatally in the nasal placode and migrate into the forebrain along the olfactory-vomeronasal nerves. Alterations in this migratory process lead to defective GnRH-1 secretion, resulting in heterogeneous genetic disorders such as idiopathic hypogonadotropic hypogonadism (IHH), and other reproductive diseases characterized by the reduction or failure of sexual competence. Combining mouse genetics with in vitro models, we demonstrate that Semaphorin 7A (Sema7A) is essential for the development of the GnRH-1 neuronal system. Loss of Sema7A signaling alters the migration of GnRH-1 neurons, resulting in significantly reduced numbers of these neurons in the adult brain as well as in reduced gonadal size and subfertility. We also show that GnRH-1 cells differentially express the Sema7 receptors ß1-integrin and Plexin C1 as a function of their migratory stage, whereas the ligand is robustly expressed along developing olfactory/vomeronasal fibers. Disruption of Sema7A function in vitro inhibits ß1-integrin-mediated migration. Analysis of Plexin C1(-/-) mice did not reveal any difference in the migratory process of GnRH-1 neurons, indicating that Sema7A mainly signals through ß1-integrin to regulate GnRH-1 cell motility. In conclusion, we have identified Sema7A as a gene implicated in the normal development of the GnRH-1 system in mice and as a genetic marker for the elucidation of some forms of GnRH-1 deficiency in humans.


Asunto(s)
Antígenos CD/metabolismo , Movimiento Celular , Fertilidad , Hormona Liberadora de Gonadotropina/metabolismo , Gónadas/embriología , Integrina beta1/metabolismo , Precursores de Proteínas/metabolismo , Semaforinas/metabolismo , Transducción de Señal , Animales , Axones/metabolismo , Encéfalo/embriología , Encéfalo/patología , Recuento de Células , Gónadas/anomalías , Gónadas/metabolismo , Gónadas/patología , Humanos , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Bulbo Olfatorio/embriología , Bulbo Olfatorio/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/deficiencia , Testículo/embriología , Testículo/metabolismo , Testículo/patología , Órgano Vomeronasal/embriología , Órgano Vomeronasal/metabolismo
12.
J Immunol ; 186(5): 2881-8, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21270411

RESUMEN

Basophils are strong mediators of Th2 responses during helminthic infections. Recently, basophils were shown to function as APCs and promote both Th2 skewing and humoral memory responses. However, the mechanisms that regulate basophils are still unclear. In this article, we show that a class IV semaphorin, Sema4B, negatively regulates basophil functions through T cell-basophil contacts. In a screen to identify semaphorins that function in the immune system, we determined that Sema4B is expressed in T and B cells. Interestingly, Sema4B(-/-) mice had considerably increased serum IgE levels despite normal lymphocyte and dendritic cell functions. Recombinant Sema4B significantly inhibited IL-4 and IL-6 production from basophils in response to various stimuli, including IL-3, papain, and FcεRI cross-linking. In addition, T cell-derived Sema4B, which accumulated at contact sites between basophils and CD4(+) T cells, suppressed basophil-mediated Th2 skewing, suggesting that Sema4B regulates basophil responses through cognate cell-cell contacts. Furthermore, Sema4B(-/-) mice had enhanced basophil-mediated memory IgE production, which was abolished by treating with an anti-FcεRIα Ab. Collectively, these results indicate that Sema4B negatively regulates basophil-mediated Th2 and humoral memory responses.


Asunto(s)
Basófilos/inmunología , Basófilos/metabolismo , Regulación hacia Abajo/inmunología , Tolerancia Inmunológica , Semaforinas/fisiología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/sangre , Memoria Inmunológica/genética , Interleucina-4/antagonistas & inhibidores , Interleucina-4/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Semaforinas/deficiencia , Semaforinas/genética , Semaforinas/aislamiento & purificación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Th2/citología , Células Th2/inmunología
13.
J Immunol ; 184(3): 1499-506, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20038643

RESUMEN

Although semaphorins were originally identified as axonal guidance molecules during neuronal development, it is emerging that several semaphorins play crucial roles in various phases of immune responses. Sema4D/CD100, a class IV semaphorin, has been shown to be involved in the nervous and immune systems through its receptors plexin-B1 and CD72, respectively. However, the involvement of Sema4D in neuroinflammation still remains unclear. We found that Sema4D promoted inducible NO synthase expression by primary mouse microglia, the effects of which were abolished in plexin-B1-deficient but not in CD72-deficient microglia. In addition, during the development of experimental autoimmune encephalomyelitis (EAE), which was induced by immunization with myelin oligodendrocyte glycoprotein-derived peptides, we observed that the expression of Sema4D and plexin-B1 was induced in infiltrating mononuclear cells and microglia, respectively. Consistent with these expression profiles, when myelin oligodendrocyte glycoprotein-specific T cells derived from wild-type mice were adoptively transferred into plexin-B1-deficient mice or bone marrow chimera mice with plexin-B1-deficient CNS resident cells, the development of EAE was considerably attenuated. Furthermore, blocking Abs against Sema4D significantly inhibited neuroinflammation during EAE development. Collectively, our findings demonstrate the role of Sema4D-plexin-B1 interactions in the activation of microglia and provide their pathologic significance in neuroinflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Microglía/inmunología , Microglía/metabolismo , Proteínas del Tejido Nervioso/fisiología , Receptores de Superficie Celular/fisiología , Semaforinas/fisiología , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/enzimología , Microglía/patología , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Quimera por Radiación/inmunología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Semaforinas/deficiencia , Semaforinas/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
14.
Nature ; 446(7136): 680-4, 2007 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-17377534

RESUMEN

Semaphorins are axon guidance factors that assist growing axons in finding appropriate targets and forming synapses. Emerging evidence suggests that semaphorins are involved not only in embryonic development but also in immune responses. Semaphorin 7A (Sema7A; also known as CD108), which is a glycosylphosphatidylinositol-anchored semaphorin, promotes axon outgrowth through beta1-integrin receptors and contributes to the formation of the lateral olfactory tract. Although Sema7A has been shown to stimulate human monocytes, its function as a negative regulator of T-cell responses has also been reported. Thus, the precise function of Sema7A in the immune system remains unclear. Here we show that Sema7A, which is expressed on activated T cells, stimulates cytokine production in monocytes and macrophages through alpha1beta1 integrin (also known as very late antigen-1) as a component of the immunological synapse, and is critical for the effector phase of the inflammatory immune response. Sema7A-deficient (Sema7a-/-) mice are defective in cell-mediated immune responses such as contact hypersensitivity and experimental autoimmune encephalomyelitis. Although antigen-specific and cytokine-producing effector T cells can develop and migrate into antigen-challenged sites in Sema7a-/- mice, Sema7a-/- T cells fail to induce contact hypersensitivity even when directly injected into the antigen-challenged sites. Thus, the interaction between Sema7A and alpha1beta1 integrin is crucial at the site of inflammation. These findings not only identify a function of Sema7A as an effector molecule in T-cell-mediated inflammation, but also reveal a mechanism of integrin-mediated immune regulation.


Asunto(s)
Antígenos CD/metabolismo , Inflamación/inmunología , Integrina alfa1beta1/metabolismo , Semaforinas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD/genética , Citocinas/metabolismo , Inmunidad/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Semaforinas/deficiencia , Semaforinas/genética , Transducción de Señal
15.
Int Immunol ; 17(10): 1277-82, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16113236

RESUMEN

Co-receptors on the B-cell surface regulate B-cell antigen receptor (BCR) signaling; however, it remains unclear how BCR signals are coordinated to maintain immune homeostasis. CD72, a negative regulator of B-cell responses, has immunoreceptor tyrosine-based inhibitory motifs within its cytoplasmic region, and the tyrosine phosphatase SHP-1 binds these sites. The natural ligand of CD72, CD100/Sema4D, belongs to the semaphorin family and induces the dissociation of SHP-1 from CD72, thereby switching off the negative signals of CD72. In the absence of CD100, BCR signals are significantly suppressed due to the constitutive association of SHP-1 with CD72, resulting in B-cell hyporesponsiveness. Here we show that CD100 regulates the sensitivity of the BCR by preventing the association of the CD72 with BCR, and this interaction is required for proper B-cell homeostasis. Consequently, as CD100-deficient mice age, they accumulate marginal zone B cells and develop high auto-antibody levels and autoimmunity. Collectively, our findings indicate that the strength of BCR signals is strictly tuned by the interaction of CD100 with CD72, and this interaction is essential for maintaining immunological homeostasis as well as generating a proper immune response.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/inmunología , Homeostasis/inmunología , Receptores de Antígenos de Linfocitos B/fisiología , Semaforinas/metabolismo , Transducción de Señal/inmunología , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos B/fisiología , Células Cultivadas , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Semaforinas/deficiencia , Semaforinas/genética , Semaforinas/fisiología , Transducción de Señal/genética
16.
Nat Cell Biol ; 6(12): 1204-11, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15543137

RESUMEN

Cardiac chamber formation involves dynamic changes in myocardial organization, including trabeculation and expansion of the compact layer. The positional cues that regulate myocardial patterning, however, remain unclear. Through ligation of the Plexin-A1 receptor, the transmembrane-type semaphorin Sema6D regulates endocardial cell migration. Here, we demonstrate that knockdown of either Sema6D or Plexin-A1 leads to the generation of a small, thin ventricular compact layer and to defective trabeculation. In the heart, expression of the Plexin-A1 extracellular domain alone can rescue the defective trabeculation induced by suppression of Plexin-A1, but not that resulting from defective Sema6D expression. This indicates that reverse signalling by Sema6D occurs within the myocardium. In a ligand-dependent manner, Abl kinase is recruited to the cytoplasmic tail of Sema6D and activated, resulting in phosphorylation of Enabled and dissociation from Sema6D. Constitutive activation of Sema6D signalling enhances the migration of myocardial cells into the trabeculae, whereas inhibition arrests cells within the compact layer. Thus, Sema6D coordinates both compact-layer expansion and trabeculation, functioning as both a ligand and a receptor for Plexin-A1.


Asunto(s)
Cardiopatías Congénitas/metabolismo , Corazón/embriología , Miocardio/metabolismo , Organogénesis/fisiología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Semaforinas/deficiencia , Semaforinas/metabolismo , Animales , Movimiento Celular/genética , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Cardiopatías Congénitas/genética , Humanos , Ligandos , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas v-abl/genética , Proteínas Oncogénicas v-abl/metabolismo , Fosforilación , Estructura Terciaria de Proteína/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/aislamiento & purificación , Semaforinas/genética , Semaforinas/aislamiento & purificación , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA