Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nature ; 626(7997): 128-135, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38233523

RESUMEN

The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.


Asunto(s)
Empalme Alternativo , Receptores Acoplados a Proteínas G , Receptores de Péptidos , Sinapsis , Animales , Ratones , Empalme Alternativo/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gs , Ligandos , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Sinapsis/metabolismo , Transducción de Señal
2.
FASEB J ; 35(5): e21515, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811688

RESUMEN

The conserved endoplasmic reticulum (ER) membrane protein TRAPα (translocon-associated protein, also known as signal sequence receptor 1, SSR1) has been reported to play a critical but unclear role in insulin biosynthesis. TRAPα/SSR1 is one component of a four-protein complex including TRAPß/SSR2, TRAPγ/SSR3, and TRAPδ/SSR4. The TRAP complex topologically has a small exposure on the cytosolic side of the ER via its TRAPγ/SSR3 subunit, whereas TRAPß/SSR2 and TRAPδ/SSR4 function along with TRAPα/SSR1 largely on the luminal side of the ER membrane. Here, we have examined pancreatic ß-cells with deficient expression of either TRAPß/SSR2 or TRAPδ/SSR4, which does not perturb mRNA expression levels of other TRAP subunits, or insulin mRNA. However, deficient protein expression of TRAPß/SSR2 and, to a lesser degree, TRAPδ/SSR4, diminishes the protein levels of other TRAP subunits, concomitant with deficient steady-state levels of proinsulin and insulin. Deficient TRAPß/SSR2 or TRAPδ/SSR4 is not associated with any apparent defect of exocytotic mechanism but rather by a decreased abundance of the proinsulin and insulin that accompanies glucose-stimulated secretion. Amino acid pulse labeling directly establishes that much of the steady-state deficiency of intracellular proinsulin can be accounted for by diminished proinsulin biosynthesis, observed in a pulse-labeling as short as 5 minutes. The proinsulin and insulin levels in TRAPß/SSR2 or TRAPδ/SSR4 null mutant ß-cells are notably recovered upon re-expression of the missing TRAP subunit, accompanying a rebound of proinsulin biosynthesis. Remarkably, overexpression of TRAPα/SSR1 can also suppress defects in ß-cells with diminished expression of TRAPß/SSR2, strongly suggesting that TRAPß/SSR2 is needed to support TRAPα/SSR1 function.


Asunto(s)
Proteínas de Unión al Calcio/deficiencia , Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Insulina/biosíntesis , Insulinoma/patología , Glicoproteínas de Membrana/deficiencia , Proinsulina/biosíntesis , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores de Péptidos/deficiencia , Animales , Células Cultivadas , Células Secretoras de Insulina/citología , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas
3.
Int J Mol Sci ; 21(21)2020 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-33147779

RESUMEN

Hyaline fibromatosis syndrome (HFS), resulting from ANTXR2 mutations, is an ultra-rare disease that causes intestinal lymphangiectasia and protein-losing enteropathy (PLE). The mechanisms leading to the gastrointestinal phenotype in these patients are not well defined. We present two patients with congenital diarrhea, severe PLE and unique clinical features resulting from deleterious ANTXR2 mutations. Intestinal organoids were generated from one of the patients, along with CRISPR-Cas9 ANTXR2 knockout, and compared with organoids from two healthy controls. The ANTXR2-deficient organoids displayed normal growth and polarity, compared to controls. Using an anthrax-toxin assay we showed that the c.155C>T mutation causes loss-of-function of ANTXR2 protein. An intrinsic defect of monolayer formation in patient-derived or ANTXR2KO organoids was not apparent, suggesting normal epithelial function. However, electron microscopy and second harmonic generation imaging showed abnormal collagen deposition in duodenal samples of these patients. Specifically, collagen VI, which is known to bind ANTXR2, was highly expressed in the duodenum of these patients. In conclusion, despite resistance to anthrax-toxin, epithelial cell function, and specifically monolayer formation, is intact in patients with HFS. Nevertheless, loss of ANTXR2-mediated signaling leads to collagen VI accumulation in the duodenum and abnormal extracellular matrix composition, which likely plays a role in development of PLE.


Asunto(s)
Colágeno/metabolismo , Duodeno/metabolismo , Síndrome de Fibromatosis Hialina/metabolismo , Enteropatías Perdedoras de Proteínas/metabolismo , Receptores de Péptidos/genética , Antígenos Bacterianos/química , Toxinas Bacterianas/química , Sistemas CRISPR-Cas , Consanguinidad , Diarrea/congénito , Matriz Extracelular/metabolismo , Humanos , Síndrome de Fibromatosis Hialina/genética , Lactante , Masculino , Microscopía Electrónica , Mutación , Fenotipo , Enteropatías Perdedoras de Proteínas/genética , Receptores de Péptidos/deficiencia , Transducción de Señal
4.
Nature ; 501(7465): 63-8, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23995686

RESUMEN

Bacillus anthracis, the causative agent of anthrax disease, is lethal owing to the actions of two exotoxins: anthrax lethal toxin (LT) and oedema toxin (ET). The key tissue targets responsible for the lethal effects of these toxins are unknown. Here we generated cell-type-specific anthrax toxin receptor capillary morphogenesis protein-2 (CMG2)-null mice and cell-type-specific CMG2-expressing mice and challenged them with the toxins. Our results show that lethality induced by LT and ET occurs through damage to distinct cell types; whereas targeting cardiomyocytes and vascular smooth muscle cells is required for LT-induced mortality, ET-induced lethality occurs mainly through its action in hepatocytes. Notably, and in contradiction to what has been previously postulated, targeting of endothelial cells by either toxin does not seem to contribute significantly to lethality. Our findings demonstrate that B. anthracis has evolved to use LT and ET to induce host lethality by coordinately damaging two distinct vital systems.


Asunto(s)
Antígenos Bacterianos/toxicidad , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/toxicidad , Animales , Carbunco/genética , Carbunco/metabolismo , Carbunco/microbiología , Resistencia a la Enfermedad/genética , Edema/inducido químicamente , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Especificidad de Órganos/efectos de los fármacos , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Especificidad por Sustrato/efectos de los fármacos , Análisis de Supervivencia
5.
Proc Natl Acad Sci U S A ; 113(8): 2294-9, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26842836

RESUMEN

Rupture of the ovarian follicle releases the oocyte at ovulation, a timed event that is critical for fertilization. It is not understood how the protease activity required for rupture is directed with precise timing and localization to the outer surface, or apex, of the follicle. We hypothesized that vasoconstriction at the apex is essential for rupture. The diameter and blood flow of individual vessels and the thickness of the apical follicle wall were examined over time to expected ovulation using intravital multiphoton microscopy. Vasoconstriction of apical vessels occurred within hours preceding follicle rupture in wild-type mice, but vasoconstriction and rupture were absent in Amhr2(cre/+)SmoM2 mice in which follicle vessels lack the normal association with vascular smooth muscle. Vasoconstriction is not simply a response to reduced thickness of the follicle wall; vasoconstriction persisted in wild-type mice when thinning of the follicle wall was prevented by infusion of protease inhibitors into the ovarian bursa. Ovulation was inhibited by preventing the periovulatory rise in the expression of the vasoconstrictor endothelin 2 by follicle cells of wild-type mice. In these mice, infusion of vasoconstrictors (either endothelin 2 or angiotensin 2) into the bursa restored the vasoconstriction of apical vessels and ovulation. Additionally, infusion of endothelin receptor antagonists into the bursa of wild-type mice prevented vasoconstriction and follicle rupture. Processing tissue to allow imaging at increased depth through the follicle and transabdominal ultrasonography in vivo showed that decreased blood flow is restricted to the apex. These results demonstrate that vasoconstriction at the apex of the follicle is essential for ovulation.


Asunto(s)
Folículo Ovárico/irrigación sanguínea , Folículo Ovárico/fisiología , Ovulación/fisiología , Vasoconstricción/fisiología , Animales , Endotelina-2/deficiencia , Endotelina-2/genética , Endotelina-2/fisiología , Femenino , Microscopía Intravital , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Folículo Ovárico/diagnóstico por imagen , Ovulación/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptor Smoothened , Ultrasonografía , Vasoconstricción/genética
6.
J Immunoassay Immunochem ; 38(1): 21-33, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27379831

RESUMEN

The failure of therapies targeting tumor angiogenesis may be caused by anti-angiogenic resistance mechanisms induced by VEGF and non-VEGF pathways alterations. Anti-angiogenic therapy failure is also attributed to immune system, acting by tumor-associated macrophages that release pro-angiogenic factors and a consequent increase of blood vessels. Recently, in a study by Rheal et al., a new angiogenic receptor, epidermal growth factor, latrophilin, and 7 trans-membrane domain-containing protein 1 on chromosome 1(ELTD1) has been identified as a promising glioma biomarker. In this study we aim to analyse whether this receptor may be used as a target molecule in glioblastoma therapy. Our results showed that small interfering RNA silencing ELTD1 caused cytotoxicity in glioblastoma cells. We also found that PDGFR, VEGFR, and their common PI3K/mTOR intracellular pathway inactivation-induced cytotoxicity in glioblastoma cells. Further, we found high percent of cytotoxicity in a low passage glioblastoma cell line after BEZ235 (a dual inhibitor of PI3K/mTOR pathway) treatment at nanomolar concentrations, compared to AG1433 (a PDGFR inhibitor) and SU1498 (a VEGFR inhibitor) that were only cytotoxic at micromolar ranges. In the future, these could prove as attractive therapeutic targets in single therapy or coupled with classic therapeutic approaches such as chemotherapy of radiotherapy.


Asunto(s)
Factor de Crecimiento Epidérmico/deficiencia , Silenciador del Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Péptidos/deficiencia , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Muerte Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Silenciador del Gen/efectos de los fármacos , Glioblastoma/genética , Humanos , ARN Interferente Pequeño/farmacología , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/biosíntesis , Receptores de Péptidos/genética
7.
Development ; 138(10): 1967-75, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490063

RESUMEN

During male sexual differentiation, the transforming growth factor-ß (TGF-ß) signaling molecule anti-Müllerian hormone (AMH; also known as Müllerian inhibiting substance, MIS) is secreted by the fetal testes and induces regression of the Müllerian ducts, the primordia of the female reproductive tract organs. Currently, the molecular identity of downstream events regulated by the AMH signaling pathway remains unclear. We found that male-specific Wnt4 expression in mouse Müllerian duct mesenchyme depends upon AMH signaling, implicating the WNT pathway as a downstream mediator of Müllerian duct regression. Inactivation of ß-catenin, a mediator of the canonical WNT pathway, did not affect AMH signaling activation in the Müllerian duct mesenchyme, but did block Müllerian duct regression. These data suggest that ß-catenin mediates AMH signaling for Müllerian duct regression during male sexual differentiation.


Asunto(s)
Conductos Paramesonéfricos/embriología , Diferenciación Sexual/fisiología , Testículo/embriología , beta Catenina/fisiología , Animales , Hormona Antimülleriana/fisiología , Secuencia de Bases , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Conductos Paramesonéfricos/metabolismo , Embarazo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Diferenciación Sexual/genética , Transducción de Señal , Testículo/metabolismo , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/fisiología , Proteína Wnt4 , beta Catenina/deficiencia , beta Catenina/genética
8.
J Lipid Res ; 54(1): 85-96, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23081987

RESUMEN

A high-fat diet (HFD) is a well-known contributing factor in the development of obesity. Most rats fed HFDs become obese. Those that avoid obesity when fed HFDs are considered diet resistant (DR). We performed a microarray screen to identify genes specific to the mesenteric fat of DR rats and revealed high expression of guanylin and guanylyl cyclase C (GC-C) in some subjects. Our histologic studies revealed that the cellular source of guanylin and GC-C is macrophages. Therefore, we developed double-transgenic (Tg) rats overexpressing guanylin and GC-C in macrophages and found that they were resistant to the effects of HFDs. In the mesenteric fat of HFD-fed Tg rats, Fas and perilipin mRNAs were downregulated, and those of genes involved in fatty acid oxidation were upregulated, compared with the levels in HFD-fed wild-type rats. In vitro studies demonstrated that lipid accumulation was markedly inhibited in adipocytes cocultured with macrophages expressing guanylin and GC-C and that this inhibition was reduced after treatment with guanylin- and GC-C-specific siRNAs. Our results suggest that the macrophagic guanylin-GC-C system contributes to the altered expression of genes involved in lipid metabolism, leading to resistance to obesity.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hormonas Gastrointestinales/metabolismo , Macrófagos/metabolismo , Mesenterio/citología , Péptidos Natriuréticos/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/metabolismo , Adipocitos/metabolismo , Animales , Colesterol/sangre , Ácidos Grasos no Esterificados/sangre , Hormonas Gastrointestinales/deficiencia , Hormonas Gastrointestinales/genética , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Insulina/sangre , Hígado/metabolismo , Macrófagos/enzimología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/metabolismo , Masculino , Péptidos Natriuréticos/deficiencia , Péptidos Natriuréticos/genética , Oxidación-Reducción , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Transgénicas , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/deficiencia , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Triglicéridos/sangre , Triglicéridos/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(17): 7203-8, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19359476

RESUMEN

Many behavioral traits and most brain disorders are common to males and females but are more evident in one sex than the other. The control of these subtle sex-linked biases is largely unstudied and has been presumed to mirror that of the highly dimorphic reproductive nuclei. Sexual dimorphism in the reproductive tract is a product of Müllerian inhibiting substance (MIS), as well as the sex steroids. Males with a genetic deficiency in MIS signaling are sexually males, leading to the presumption that MIS is not a neural regulator. We challenge this presumption by reporting that most immature neurons in mice express the MIS-specific receptor (MISRII) and that male Mis(-/-) and Misrii(-/-) mice exhibit subtle feminization of their spinal motor neurons and of their exploratory behavior. Consequently, MIS may be a broad regulator of the subtle sex-linked biases in the nervous system.


Asunto(s)
Hormona Antimülleriana/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo , Caracteres Sexuales , Andrógenos/metabolismo , Animales , Hormona Antimülleriana/deficiencia , Hormona Antimülleriana/genética , Encéfalo/embriología , Proliferación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Columna Vertebral/citología , Columna Vertebral/metabolismo
10.
J Neurochem ; 113(6): 1644-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20374426

RESUMEN

Lesioned axons do not regenerate in the adult mammalian CNS, owing to the over-expression of inhibitory molecules such as myelin-derived proteins or chondroitin sulphate proteoglycans. In order to overcome axon inhibition, strategies based on extrinsic and intrinsic treatments have been developed. For myelin-associated inhibition, blockage with NEP1-40, receptor bodies or IN-1 antibodies has been used. In addition, endogenous blockage of cell signalling mechanisms induced by myelin-associated proteins is a potential tool for overcoming axon inhibitory signals. We examined the participation of glycogen synthase kinase 3beta (GSK3beta) and extracellular-related kinase (ERK) 1/2 in axon regeneration failure in lesioned cortical neurons. We also investigated whether pharmacological blockage of GSK3beta and ERK1/2 activities facilitates regeneration after myelin-directed inhibition in two models: (i) cerebellar granule cells and (ii) lesioned entorhino-hippocampal pathway in slice cultures, and whether the regenerative effects are mediated by Nogo Receptor 1 (NgR1). We demonstrate that, in contrast to ERK1/2 inhibition, the pharmacological treatment of GSK3beta inhibition strongly facilitated regrowth of cerebellar granule neurons over myelin independently of NgR1. Finally, these regenerative effects were corroborated in the lesioned entorhino-hippocampal pathway in NgR1-/- mutant mice. These results provide new findings for the development of new assays and strategies to enhance axon regeneration in injured cortical connections.


Asunto(s)
Corteza Cerebral/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Neuritas/fisiología , Neuronas/citología , Aminofenoles/farmacología , Animales , Animales Recién Nacidos , Axotomía/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteínas Ligadas a GPI , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/citología , Humanos , Indoles/farmacología , Lisina/análogos & derivados , Lisina/metabolismo , Maleimidas/farmacología , Ratones , Ratones Noqueados , Mutación/genética , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/farmacología , Neuritas/efectos de los fármacos , Proteínas Nogo , Receptor Nogo 1 , Embarazo , Ratas , Receptores de Superficie Celular , Receptores de Péptidos/deficiencia , Factores de Tiempo , Versicanos/metabolismo
11.
J Exp Med ; 189(4): 657-62, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9989980

RESUMEN

N-formylpeptides derive from bacterial and mitochondrial proteins, and bind to specific receptors on mammalian phagocytes. Since binding induces chemotaxis and activation of phagocytes in vitro, it has been postulated that N-formylpeptide receptor signaling in vivo may be important in antimicrobial host defense, although direct proof has been lacking. Here we test this hypothesis in mice lacking the high affinity N-formylpeptide receptor (FPR), created by targeted gene disruption. FPR-/- mice developed normally, but had increased susceptibility to challenge with Listeria monocytogenes, as measured by increased mortality compared with wild-type littermates. FPR-/- mice also had increased bacterial load in spleen and liver 2 d after infection, which is before development of a specific cellular immune response, suggesting a defect in innate immunity. Consistent with this, neutrophil chemotaxis in vitro and neutrophil mobilization into peripheral blood in vivo in response to the prototype N-formylpeptide fMLF (formyl-methionyl-leucyl-phenylalanine) were both absent in FPR-/- mice. These results indicate that FPR functions in antibacterial host defense in vivo.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Listeriosis/inmunología , Receptores Inmunológicos/fisiología , Receptores de Péptidos/fisiología , Animales , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/genética , Predisposición Genética a la Enfermedad , Inmunidad Innata , Recuento de Leucocitos , Listeria monocytogenes/aislamiento & purificación , Listeriosis/microbiología , Hígado/microbiología , Ratones , Ratones Noqueados , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/fisiología , Receptores de Formil Péptido , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Bazo/microbiología
12.
Mol Cell Biol ; 26(20): 7760-71, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17015483

RESUMEN

Translocon-associated protein complex (TRAP) is thought to be required for efficient protein-specific translocation across the endoplasmic reticulum membrane. We created a mutation in the Trapalpha gene that leads to the synthesis of a truncated TRAPalpha protein fused to ShBle-beta-galactosidase. Analysis of Trapalpha cDNAs reveals that among three different messenger RNAs expressed in the mouse, one of them encodes a slightly larger protein that differs in its C-terminal end. This mRNA, specific for skeletal muscle and heart, is only expressed after birth. Homozygous Trapalpha mutant pups die at birth, likely as a result of severe cardiac defects. Indeed, the septation of the proximal part of the outflow tract is absent, resulting in a double-outlet right ventricle. Studies of protein secretion in transfected embryonic fibroblasts reveal that the TRAP complex does not function properly in homozygous mutant cells and confirm, in vivo, the involvement of TRAP in substrate-specific translocation. Our results provide the first in vivo demonstration that a member of the TRAP complex plays a crucial role in mammalian heart development and suggest that TRAPalpha could be involved in translocation of factors necessary for maturation of endocardial cushions.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Péptidos/metabolismo , Envejecimiento/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Movimiento Celular , Secuencia Conservada , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Corazón/embriología , Homocigoto , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Miocardio/metabolismo , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Péptidos/química , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia
13.
Reprod Fertil Dev ; 21(3): 440-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19261221

RESUMEN

The peptide hormone relaxin is expressed in the prostate gland and secreted into the seminal plasma; however, its function within the prostate has not been established. Relaxin-mutant mice (Rln(-/-)) were reported to have abnormal prostate morphology, but there was no prostate phenotype in relaxin receptor-mutant (Rxfp1(-/-)) mice. The present study aimed to verify the phenotypes in the anterior, dorsal and lateral lobes of the prostate gland of Rln(-/-) and Rxfp1(-/-) mice at different adult ages. Rln(-/-) mice were also treated with relaxin to evaluate the effects of exogenously administered hormone on prostate morphology. Comparisons between these three lobes of the prostate demonstrated no obvious differences in duct morphology, epithelial height or collagen density between Rln(+/+) and Rln(-/-) mice at 2, 4, 6, 8 and 12 months of age. This was similar in Rxfp1(-/-) mice. Relaxin treatment did not affect morphology or epithelial cell height in the different lobes. Furthermore, prostate lobe morphology in transgenic mice overexpressing relaxin Tg(Rln) was not different from the wild-type controls. Rxfp1 was detected in the prostate throughout adult life, but there was no consistent expression of relaxin. In summary, the present study found no evidence to support a prostate phenotype in adult Rln- or Rxfp1-mutant mice.


Asunto(s)
Próstata/anatomía & histología , Relaxina/deficiencia , Envejecimiento , Animales , Células Epiteliales/citología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Próstata/química , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Relaxina/genética , Relaxina/farmacología
15.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29863500

RESUMEN

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Asunto(s)
Inmunoconjugados/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Brentuximab Vedotina , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Proteínas de Microfilamentos , Neoplasias/metabolismo , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Células del Estroma/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Neurosci ; 26(47): 12242-50, 2006 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-17122049

RESUMEN

The hypothesis that Nogo-A (Reticulon 4A) and Nogo-66 receptor (NgR1) limit adult CNS axonal growth after injury is supported by both in vitro experiments and in vivo pharmacological studies. However, genetic assessment of the role of Nogo-A in corticospinal tract (CST) axons after spinal cord dorsal hemisection has yielded conflicting results. CST regeneration is detected in homozygous nogo-ab(trap/trap) mice, but not in nogo-ab(atg/atg) mice. CST regeneration is also present after pharmacological NgR blockade, but not in ngr1(-/-) mice. To assess the nogo-ab(atg) and ngr1-null alleles for other axon growth phenotypes, we created unilateral pyramidotomies and monitored the uninjured CST. There is robust pyramidotomy-induced growth of nogo-ab(atg/atg) and ngr1(-/-) CST axons into denervated cervical gray matter. This fiber growth correlates with recovery of fine motor skill in the affected forelimb. Thus nogo-ab and ngr1 play a modulated role in limiting CNS axonal growth across a spectrum of different tracts in various lesion models.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/citología , Proteínas de la Mielina/metabolismo , Receptores de Péptidos/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Conducta Animal , Péptido Relacionado con Gen de Calcitonina/metabolismo , Lateralidad Funcional , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Básica de Mielina/metabolismo , Proteínas Nogo , Proteína Quinasa C/metabolismo , Desempeño Psicomotor/fisiología , Tractos Piramidales/lesiones , Tractos Piramidales/metabolismo , Tractos Piramidales/fisiología , Receptores de Péptidos/deficiencia
17.
Eur J Neurosci ; 26(12): 3339-44, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18052978

RESUMEN

New neurons are added on a daily basis to the olfactory bulb (OB) of a mammal, and this phenomenon exists throughout its lifetime. These new cells are born in the subventricular zone and migrate to the OB via the rostral migratory stream (RMS). To examine the role of the prokineticin receptor 2 (Prokr2) in neurogenesis, we created a Prokr2 null mouse, and report a decrease in the volume of its OB and also a decrease in the number of bromodeoxyuridine (BrdU)-positive cells. There is disrupted architecture of the OB, with the glomerular layer containing terminal dUTP nick-end labeling (TUNEL) -positive nuclei and also a decrease in tyrosine hydroxylase-positive neurons in this layer. In addition, there are increased numbers of doublecortin-positive neuroblasts in the RMS and increased PSA-NCAM (polysialylated form of the neural cell adhesion molecule) -positive neuronal progenitors around the olfactory ventricle, indicating their detachment from homotypic chains is compromised. Finally, in support of this, Prokr2-deficient cells expanded in vitro as neurospheres are incapable of migrating towards a source of recombinant human prokineticin 2 (PROK2). Together, these findings suggest an important role for Prokr2 in OB neurogenesis.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Neuronas/patología , Bulbo Olfatorio/anomalías , Bulbo Olfatorio/patología , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Péptidos/deficiencia , Células Madre/patología , Animales , Animales Recién Nacidos , Bromodesoxiuridina , Recuento de Células , Movimiento Celular/efectos de los fármacos , Proteínas de Dominio Doblecortina , Femenino , Hormonas Gastrointestinales/farmacología , Homocigoto , Humanos , Lactancia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/farmacología , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/fisiopatología , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Proteínas Recombinantes/farmacología , Ácidos Siálicos/metabolismo , Esferoides Celulares , Células Madre/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
18.
Mol Cell Biol ; 23(14): 4882-91, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12832474

RESUMEN

The inhibins are gonadal transforming growth factor beta superfamily protein hormones that suppress pituitary follicle-stimulating hormone (FSH) synthesis. Recently, betaglycan and inhibin binding protein (InhBP/p120, also known as the product of immunoglobulin superfamily gene 1 [IGSF1]) were identified as candidate inhibin coreceptors, shedding light on the molecular basis of how inhibins may affect target cells. Activins, which are structurally related to the inhibins, act within the pituitary to stimulate FSH production. Betaglycan increases the affinity of inhibins for the activin type IIA (ACVR2) receptor, thereby blocking activin binding and signaling through this receptor. InhBP/p120 may not directly bind inhibins but may interact with the activin type IB receptor, ALK4, and participate in inhibin B's antagonism of activin signaling. To better understand the in vivo functions of InhBP/p120, we characterized the InhBP/p120 mRNAs and gene in mice and generated InhBP/p120 mutant mice by gene targeting in embryonic stem cells. InhBP/p120 mutant male and female mice were viable and fertile. Moreover, they showed no alterations in FSH synthesis or secretion or in ovarian or testicular function. These data contribute to a growing body of evidence indicating that InhBP/p120 does not play an essential role in inhibin biology.


Asunto(s)
Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Reproducción/fisiología , Animales , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante de Subunidad beta/sangre , Hormona Folículo Estimulante de Subunidad beta/genética , Expresión Génica , Marcación de Gen , Genotipo , Gonadotropinas/sangre , Masculino , Ratones , Ratones Mutantes , Fenotipo , Hipófisis/citología , Hipófisis/fisiología , Isoformas de Proteínas , ARN Mensajero/metabolismo , Receptores de Péptidos/deficiencia
19.
Ann N Y Acad Sci ; 1041: 82-90, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15956690

RESUMEN

We studied ligand-receptor interactions between relaxin (RLN), insulin-like 3 peptide (INSL3), and LGR7 and LGR8 receptors. The phenotypic effects of deficiency for Lgr7 and Lgr8 receptors, transgenic overexpression of Rln1 and Insl3, and different combinations of these mutations in mice were analyzed. It was reported that Rln1-deficient mice exhibit abnormal nipple development, prolonged parturition, age-related pulmonary fibrosis, and abnormalities in the testis and prostate. Mutation of Lgr8 or its cognate ligand Insl3 causes cryptorchidism. Mutant females deficient for the Lgr7 receptor have grossly undeveloped nipples and are unable to feed their progeny. Parturition is prolonged in these females, resulting in a significantly higher number of stillborn pups. Histologic analysis of Lgr7 mutant lung tissues demonstrates increased collagen accumulation and perivenular smooth muscle hypertrophy. However, Lgr7-deficient males do not exhibit abnormalities of male reproductive organs as seen in Rln1 knockout mice. Double-mutant males deficient for Lgr7 and Lgr8 have a normal prostate, suggesting that Lgr8 does not account for differences in Rln1-/- and Lgr7-/- phenotypes. We also produced mice with transgenic overexpression of Rln1 under rat insulin 2 promoter. Rln1 transgenic females exhibited increased nipple size, whereas Rln1 transgenic females deficient in Lgr7 had undeveloped nipples, indicating that Lgr7 is the only receptor for relaxin that mediates this effect. Transgenic overexpression of Rln1 does not affect gonadal descent in females, and transgenic overexpression of Insl3 does not rescue the mutant phenotype of Lgr7-deficient mice, suggesting the non-overlapping functions of two signaling pathways. In summary, our data indicate that the Insl3/Lgr8 and Rln1/Lgr7 pathways are distinct and separate in vivo. Therefore, we propose to rename Lgr8 as Insl3r (Insl3 receptor) and Lgr7 as Rlnr (relaxin receptor).


Asunto(s)
Insulina/metabolismo , Proteínas/metabolismo , Relaxina/metabolismo , Transducción de Señal , Animales , Línea Celular , Femenino , Regulación de la Expresión Génica , Marcación de Gen , Humanos , Insulina/genética , Insulina/farmacología , Ratones , Ratones Transgénicos , Pezones/anomalías , Pezones/metabolismo , Fenotipo , Proteínas/genética , Proteínas/farmacología , Ratas , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relaxina/genética
20.
Mol Endocrinol ; 17(12): 2639-46, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12933905

RESUMEN

During male development testes descend from their embryonic intraabdominal position into the scrotum. Two genes, encoding the insulin-like 3 peptide (INSL3) and the GREAT/LGR8 G protein-coupled receptor, control the differentiation of gubernaculum, the caudal genitoinguinal ligament critical for testicular descent. It was established that the INSL3 peptide activates GREAT/LGR8 receptor in vitro. Mutations of Insl3 or Great cause cryptorchidism (undescended testes) in mice. Overexpression of the transgenic Insl3 causes male-like gubernaculum differentiation, ovarian descent into lower abdominal position, and reduced fertility in females. To address the question whether Great deletion complements the mutant female phenotype caused by the Insl3 overexpression, we have produced Insl3 transgenic mice deficient for Great. Such females had a wild-type phenotype, demonstrating that Great was the only cognate receptor for Insl3 in vivo. We have established that pancreatic HIT cells, transfected with the INSL3 cDNA, produce functionally active peptide. Analysis of five INSL3 mutant variants detected in cryptorchid patients showed that P49S substitution renders functionally compromised peptide. Therefore, mutations in INSL3 might contribute to the etiology of cryptorchidism. We have also showed that synthetic insulin-like peptides (INSL4 and INSL6) were unable to activate LGR7 or GREAT/LGR8.


Asunto(s)
Proteínas/genética , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/fisiología , Secuencia de Aminoácidos , Animales , Criptorquidismo/genética , Femenino , Eliminación de Gen , Genotipo , Insulina , Tamaño de la Camada , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis , Embarazo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA