Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Development ; 144(14): 2584-2594, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619819

RESUMEN

Multiple signaling pathways guide the behavior and differentiation of both germline stem cells (GSCs) and somatic follicle stem cells (FSCs) in the Drosophila germarium, necessitating careful control of signal generation, range and responses. Signal integration involves escort cells (ECs), which promote differentiation of the GSC derivatives they envelop, provide niche signals for FSCs and derive directly from FSCs in adults. Hedgehog (Hh) signaling induces the Hippo pathway effector Yorkie (Yki) to promote proliferation and maintenance of FSCs, but Hh also signals to ECs, which are quiescent. Here, we show that in ECs both Hh and Yki limit production of BMP ligands to allow germline differentiation. Loss of Yki produced a more severe germarial phenotype than loss of Hh signaling and principally induced a different BMP ligand. Moreover, Yki activity reporters and epistasis tests showed that Yki does not mediate the key actions of Hh signaling in ECs. Thus, both the coupling and output of the Hh and Yki signaling pathways differ between FSCs and ECs despite their proximity and the fact that FSCs give rise directly to ECs.


Asunto(s)
Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/metabolismo , Ovario/citología , Ovario/metabolismo , Transactivadores/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Genes de Insecto , Proteínas Hedgehog/genética , Proteínas Nucleares/genética , Oogénesis/genética , Oogénesis/fisiología , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Transducción de Señal , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Nicho de Células Madre , Células Madre/citología , Células Madre/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Señalizadoras YAP
2.
J Cell Physiol ; 234(5): 6654-6666, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30341900

RESUMEN

Multiple studies have reported different methods in treating gestational diabetes mellitus (GDM); however, the relationship between miR-335-5p and GDM still remains unclear. Here, this study explores the effect of miR-335-5p on insulin resistance and pancreatic islet ß-cell secretion via activation of the TGFß signaling pathway by downregulating VASH1 expression in GDM mice. The GDM mouse model was established and mainly treated with miR-335-5p mimic, miR-335-5p inhibitor, si-VASH1, and miR-335-5p inhibitor + si-VASH1. Oral glucose tolerance test (OGTT) was conducted to detect fasting blood glucose (FBG) fasting insulin (FINS). The OGTT was also used to calculate a homeostasis model assessment of insulin resistance (HOMA-IR). A hyperglycemic clamp was performed to measure the glucose infusion rate (GIR), which estimated ß-cell function. Expressions of miR-335-5p, VASH1, TGF-ß1, and c-Myc in pancreatic islet ß-cells were determined by RT-qPCR, western blot analysis, and insulin release by ELISA. The miR-335-5p mimic and si-VASH1 groups showed elevated blood glucose levels, glucose area under the curve (GAUC), and HOMA-IR, but a reduced GIR and positive expression of VASH1. Overexpression of miR-335-5p and inhibition of VASH1 contributed to activated TGFß1 pathway, higher c-Myc, and lower VASH1 expressions, in addition to downregulated insulin and insulin release levels. These findings provided evidence that miR-335-5p enhanced insulin resistance and suppressed pancreatic islet ß-cell secretion by inhibiting VASH1, eventually activating the TGF-ß pathway in GDM mice, which provides more clinical insight on the GDM treatment.


Asunto(s)
Glucemia/genética , Diabetes Gestacional/genética , Resistencia a la Insulina/genética , MicroARNs/genética , Factor de Crecimiento Transformador beta/deficiencia , Animales , Glucemia/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa/métodos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Embarazo , Factor de Crecimiento Transformador beta/metabolismo
3.
Gen Comp Endocrinol ; 277: 122-129, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30951723

RESUMEN

Gonadal soma-derived factor (Gsdf) is a unique TGF-ß factor essential for both ovarian and testicular development in Hd-rR medaka (Oryzias latipes). However, the downstream genes regulated by Gsdf signaling remain unknown. Using a high-throughput proteomic approach, we identified a significant increase in the expression of the RNA-binding protein Igf2bp3 in gsdf-deficient ovaries. We verified this difference in transcription and protein expression against normal gonads using real-time PCR quantification and Western blotting. The genomic structure of igf2bp3 and the syntenic flanking segments are highly conserved across fish and mammals. igf2bp3 expression was correlated with oocyte development, which is consistent with the expression of the igf2bp3 ortholog Vg1-RBP/Vera in Xenopus. In contrast to the normal ovary, cysts of H3K27me3- and Igf2bp3-positive germ cells were dramatically increased in the one-month-old gsdf-deficient ovary, indicating that the gsdf depletion led to a dysregulation of Igf2bp3-mediated oocyte development. Our results provide novel insights into the Gsdf-Igf2bp3 signaling mechanisms that underlie the fundamental process of gametogenesis; these mechanisms may be well conserved across phyla.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Oryzias/genética , Proteínas de Unión al ARN/genética , Factor de Crecimiento Transformador beta/deficiencia , Secuencia de Aminoácidos , Animales , Proliferación Celular , Secuencia Conservada , Evolución Molecular , Femenino , Perfilación de la Expresión Génica , Histonas/metabolismo , Lisina/metabolismo , Masculino , Oogénesis/genética , Ovario/embriología , Ovario/metabolismo , Filogenia , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo
4.
J Neurosci ; 37(17): 4584-4592, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28363982

RESUMEN

Transforming growth factor-ß (TGF-ß) plays an important role in the development and maintenance of embryonic dopaminergic (DA) neurons in the midbrain. To study the function of TGF-ß signaling in the adult nigrostriatal system, we generated transgenic mice with reduced TGF-ß signaling in mature neurons. These mice display age-related motor deficits and degeneration of the nigrostriatal system. Increasing TGF-ß signaling in the substantia nigra through adeno-associated virus expressing a constitutively active type I receptor significantly reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration and motor deficits. These results suggest that TGF-ß signaling is critical for adult DA neuron survival and that modulating this signaling pathway has therapeutic potential in Parkinson disease.SIGNIFICANCE STATEMENT We show that reducing Transforming growth factor-ß (TGF-ß) signaling promotes Parkinson disease-related pathologies and motor deficits, and increasing TGF-ß signaling reduces neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a parkinsonism-inducing agent. Our results provide a rationale to pursue a means of increasing TGF-ß signaling as a potential therapy for Parkinson's disease.


Asunto(s)
Intoxicación por MPTP/fisiopatología , Neostriado/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Transducción de Señal , Sustancia Negra/fisiopatología , Factor de Crecimiento Transformador beta/deficiencia , Animales , Supervivencia Celular/genética , Trastornos Neurológicos de la Marcha/inducido químicamente , Trastornos Neurológicos de la Marcha/fisiopatología , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/inducido químicamente , Equilibrio Postural , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética
5.
Cell Physiol Biochem ; 42(5): 1961-1972, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28793286

RESUMEN

BACKGROUND/AIMS: Sepsis is a systemic inflammatory response during infection. There are limited therapeutic options for sepsis patients. Interleukin (IL)-33 has been reported recently with a beneficial effect in mouse sepsis. METHODS: In this study, we initiated a clinical study to measure serum levels of pro-inflammatory cytokines including IL-33 in sepsis patients. Next, we employed cecal ligation and puncture (CLP) to study the role of IL-33 during sepsis. To further dissect the molecular mechanism, we used in vivo knockout models and in vitro knockdown murine embryonic fibroblasts (MEFs) to investigate the cross-talk between IL-33 and IL-17 signaling, and to identify the potential downstream mediators. RESULTS: IL-33 and IL-17 were upregulated in both clinical and experimental sepsis. In CLP, IL-33 (-/-) mice showed higher mortality rate, and IL-33 treatment improved the survival rate. Elevated proinflammatory cytokines in sepsis were related to IL-17 from γδT cells. IL-33 treatment suppressed production of these cytokines by targeting IL-17 signaling both in vivo and in vitro. Finally, IL-33 was shown to inhibit the IL-17 pathway via activating suppressor of cytokine signaling (SOCS)-3. CONCLUSION: Collectively, the results suggest that IL-33 plays a negative regulatory role in sepsis progression by inhibiting IL-17 pathway through activating SOCS3. This finding would inspire a new therapeutic strategy for treating sepsis.


Asunto(s)
Interleucina-33/metabolismo , Receptores de Interleucina-17/metabolismo , Sepsis/diagnóstico , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , Animales , Estudios de Casos y Controles , Quimiocina CXCL1/análisis , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Interleucina-17/análisis , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-33/análisis , Interleucina-33/genética , Interleucina-6/análisis , Lentinula/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sepsis/mortalidad , Sepsis/patología , Proteína 3 Supresora de la Señalización de Citocinas/antagonistas & inhibidores , Proteína 3 Supresora de la Señalización de Citocinas/genética , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
6.
Immunity ; 28(4): 559-70, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18400195

RESUMEN

Interleukin-23 (IL-23) is an inflammatory cytokine that plays a key role in the pathogenesis of several autoimmune and inflammatory diseases. It orchestrates innate and T cell-mediated inflammatory pathways and can promote T helper 17 (Th17) cell responses. Utilizing a T cell transfer model, we showed that IL-23-dependent colitis did not require IL-17 secretion by T cells. Furthermore, IL-23-independent intestinal inflammation could develop if immunosuppressive pathways were reduced. The frequency of naive T cell-derived Foxp3+ cells in the colon increased in the absence of IL-23, indicating a role for IL-23 in controlling regulatory T cell induction. Foxp3-deficient T cells induced colitis when transferred into recipients lacking IL-23p19, showing that IL-23 was not essential for intestinal inflammation in the absence of Foxp3. Taken together, our data indicate that overriding immunosuppressive pathways is an important function of IL-23 in the intestine and could influence not only Th17 cell activity but also other types of immune responses.


Asunto(s)
Colitis/inmunología , Colitis/metabolismo , Tolerancia Inmunológica , Mediadores de Inflamación/fisiología , Interleucina-23/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Colitis/genética , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Tolerancia Inmunológica/genética , Mediadores de Inflamación/metabolismo , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-23/deficiencia , Interleucina-23/genética , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
7.
Cancer Cell ; 12(6): 514-27, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18068629

RESUMEN

The extracellular matrix (ECM) can induce chemotherapy resistance via AKT-mediated inhibition of apoptosis. Here, we show that loss of the ECM protein TGFBI (transforming growth factor beta induced) is sufficient to induce specific resistance to paclitaxel and mitotic spindle abnormalities in ovarian cancer cells. Paclitaxel-resistant cells treated with recombinant TGFBI protein show integrin-dependent restoration of paclitaxel sensitivity via FAK- and Rho-dependent stabilization of microtubules. Immunohistochemical staining for TGFBI in paclitaxel-treated ovarian cancers from a prospective clinical trial showed that morphological changes of paclitaxel-induced cytotoxicity were restricted to areas of strong expression of TGFBI. These data show that ECM can mediate taxane sensitivity by modulating microtubule stability.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Antineoplásicos Fitogénicos/farmacología , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de la Matriz Extracelular/deficiencia , Femenino , Fibronectinas/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Integrinas/metabolismo , Mitosis/efectos de los fármacos , Modelos Biológicos , Neoplasias Ováricas/patología , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Tubulina (Proteína)/metabolismo
8.
Nat Genet ; 38(12): 1424-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17099713

RESUMEN

Adult bones have a notable regenerative capacity. Over 40 years ago, an intrinsic activity capable of initiating this reparative response was found to reside within bone itself, and the term bone morphogenetic protein (BMP) was coined to describe the molecules responsible for it. A family of BMP proteins was subsequently identified, but no individual BMP has been shown to be the initiator of the endogenous bone repair response. Here we demonstrate that BMP2 is a necessary component of the signaling cascade that governs fracture repair. Mice lacking the ability to produce BMP2 in their limb bones have spontaneous fractures that do not resolve with time. In fact, in bones lacking BMP2, the earliest steps of fracture healing seem to be blocked. Although other osteogenic stimuli are still present in the limb skeleton of BMP2-deficient mice, they cannot compensate for the absence of BMP2. Collectively, our results identify BMP2 as an endogenous mediator necessary for fracture repair.


Asunto(s)
Desarrollo Óseo/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Curación de Fractura/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Desarrollo Óseo/genética , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Huesos/metabolismo , Huesos/patología , Curación de Fractura/genética , Hibridación in Situ , Ratones , Ratones Noqueados , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
9.
J Exp Med ; 204(3): 467-74, 2007 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-17353364

RESUMEN

Members of the transforming growth factor beta (TGF-beta) superfamily of growth factors have been shown to regulate the in vitro proliferation and maintenance of hematopoietic stem cells (HSCs). Working at a common level of convergence for all TGF-beta superfamily signals, Smad4 is key in orchestrating these effects. The role of Smad4 in HSC function has remained elusive because of the early embryonic lethality of the conventional knockout. We clarify its role by using an inducible model of Smad4 deletion coupled with transplantation experiments. Remarkably, systemic induction of Smad4 deletion through activation of MxCre was incompatible with survival 4 wk after induction because of anemia and histopathological changes in the colonic mucosa. Isolation of Smad4 deletion to the hematopoietic system via several transplantation approaches demonstrated a role for Smad4 in the maintenance of HSC self-renewal and reconstituting capacity, leaving homing potential, viability, and differentiation intact. Furthermore, the observed down-regulation of notch1 and c-myc in Smad4(-/-) primitive cells places Smad4 within a network of genes involved in the regulation HSC renewal.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Proteína Smad4/fisiología , Animales , Diferenciación Celular/genética , Regulación hacia Abajo/genética , Genes Letales , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Inmunofenotipificación , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/biosíntesis , Transducción de Señal/genética , Proteína Smad4/deficiencia , Proteína Smad4/genética , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
10.
Nat Genet ; 15(2): 207-11, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9020852

RESUMEN

The transforming growth factor beta 1 (TGF beta 1) signalling pathway is important in embryogenesis and has been implicated in hereditary haemorrhagic telangiectasia (HHT), atherosclerosis, tumorigenesis and immunomodulation. Therefore, identification of factors which modulate TGF beta 1 bioactivity in vivo is important. On a mixed genetic background, approximately 50% Tgfb1-/- conceptuses die midgestation from defective yolk sac vasculogenesis. The other half are developmentally normal but die three weeks postpartum. Intriguingly, the vascular defects of Tgfb1-/- mice share histological similarities to lesions seen in HHT patients. It has been suggested that dichotomy in Tgfb1-/- lethal phenotypes is due to maternal TGF beta 1 rescue of some, but not all, Tgfb1-/- embryos12. Here we show that the Tgfb1-/- phenotype depends on the genetic background of the conceptus. In NIH/Ola, C57BL/6J/Ola and F1 conceptuses, Tgfb1-/- lethality can be categorized into three developmental classes. A major codominant modifier gene of embryo lethality was mapped to proximal mouse chromosome 5, using a genome scan for non-mendelian distribution of alleles in Tgfb1-/- neonatal animals which survive prenatal lethality. This gene accounts for around three quarters of the genetic effect between mouse strains and can, in part, explain the distribution of the three lethal phenotypes. This approach, using neonatal DNA samples, is generally applicable to identification of loci that influence the effect of early embryonic lethal mutations, thus furthering knowledge of genetic interactions that occur during early mammalian development in vivo.


Asunto(s)
Muerte Fetal/genética , Genes Letales , Factor de Crecimiento Transformador beta/deficiencia , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Desarrollo Embrionario y Fetal/genética , Hematopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Repeticiones de Microsatélite , Fenotipo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/fisiología , Saco Vitelino/irrigación sanguínea
11.
Gastroenterology ; 141(5): 1887-96.e1-2, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21839042

RESUMEN

BACKGROUND & AIMS: Signaling via interleukin (IL)-10 or transforming growth factor (TGF)-ß is disrupted in subpopulations of patients with inflammatory bowel disease, but it is not clear how a T-helper (Th) 1 cell response is induced. We studied conversion of human mucosal innate immune cells into inflammatory cells and the initiation of a Th1 cell response following loss of IL-10 or TGF-ß signaling. METHODS: We depleted IL-10 or TGF-ß from explant cultures of human normal colonic mucosa using immunoneutralization. Pharmacologic inhibitors and antibodies were used to determine the factors involved in the initiation of an interferon (IFN)-γ response following loss of TGF-ß or IL-10 signaling. Cytokines produced by mucosal cells were assessed by enzyme-linked immunosorbent assay and quantitative reverse-transcriptase polymerase chain reaction. The subsets of cells involved in cytokine production were determined by in situ immunofluorescence analysis and flow cytometry after digestion of the explants with collagenase. RESULTS: Depletion of IL-10 from human normal colonic mucosa resulted in an IFN-γ response, characterized by early-stage secretion of mature IL-18 and production of the active form of caspase-1 by macrophages and some epithelial cells. A caspase-1 inhibitor or the IL-18 antagonist IL-18-binding protein blocked this response. By contrast, depletion of TGF-ß resulted in an IFN-γ response that was preceded by and required secretion of IL-12 from macrophages, dendritic cells, and epithelial cells. CONCLUSIONS: Innate immune cells (macrophages and epithelial cells) activate a Th1 cell response in explant cultures of human normal colonic mucosa depleted in IL-10 or TGF-ß via distinct, nonredundant pathways. These pathways might contribute to the pathogenesis of inflammatory bowel disease.


Asunto(s)
Colon/patología , Interleucina-10/deficiencia , Transducción de Señal/fisiología , Células TH1/patología , Factor de Crecimiento Transformador beta/deficiencia , Adulto , Anciano , Anciano de 80 o más Años , Caspasa 1/metabolismo , Células Cultivadas , Colon/metabolismo , Femenino , Humanos , Inmunidad Innata/fisiología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Células TH1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
12.
Nature ; 441(7090): 231-4, 2006 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-16648837

RESUMEN

A new lineage of effector CD4+ T cells characterized by production of interleukin (IL)-17, the T-helper-17 (T(H)17) lineage, was recently described based on developmental and functional features distinct from those of classical T(H)1 and T(H)2 lineages. Like T(H)1 and T(H)2, T(H)17 cells almost certainly evolved to provide adaptive immunity tailored to specific classes of pathogens, such as extracellular bacteria. Aberrant T(H)17 responses have been implicated in a growing list of autoimmune disorders. T(H)17 development has been linked to IL-23, an IL-12 cytokine family member that shares with IL-12 a common subunit, IL-12p40 (ref. 8). The IL-23 and IL-12 receptors also share a subunit, IL-12Rbeta1, that pairs with unique, inducible components, IL-23R and IL-12Rbeta2, to confer receptor responsiveness. Here we identify transforming growth factor-beta (TGF-beta) as a cytokine critical for commitment to T(H)17 development. TGF-beta acts to upregulate IL-23R expression, thereby conferring responsiveness to IL-23. Although dispensable for the development of IL-17-producing T cells in vitro and in vivo, IL-23 is required for host protection against a bacterial pathogen, Citrobacter rodentium. The action of TGF-beta on naive T cells is antagonized by interferon-gamma and IL-4, thus providing a mechanism for divergence of the T(H)1, T(H)2 and T(H)17 lineages.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Interleucina-17/metabolismo , Células TH1/citología , Células TH1/efectos de los fármacos , Células Th2/citología , Células Th2/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/farmacología , Interleucina-17/genética , Interleucina-17/farmacología , Interleucina-23 , Subunidad p19 de la Interleucina-23 , Interleucina-4/farmacología , Interleucinas/inmunología , Interleucinas/farmacología , Ratones , Receptores de Interleucina/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
13.
J Exp Med ; 201(5): 737-46, 2005 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-15753207

RESUMEN

CD4(+)CD25(+) regulatory T (T reg) cells play a pivotal role in control of the immune response. Transforming growth factor-beta (TGF-beta) has been shown to be required for T reg cell activity; however, precisely how it is involved in the mechanism of suppression is poorly understood. Using the T cell transfer model of colitis, we show here that CD4(+)CD45RB(high) T cells that express a dominant negative TGF-beta receptor type II (dnTbetaRII) and therefore cannot respond to TGF-beta, escape control by T reg cells in vivo. CD4(+)CD25(+) T reg cells from the thymus of dnTbetaRII mice retain the ability to inhibit colitis, suggesting that T cell responsiveness to TGF-beta is not required for the development or peripheral function of thymic-derived T reg cells. In contrast, T reg cell activity among the peripheral dnTbetaRII CD4(+)CD25(+) population is masked by the presence of colitogenic effector cells that cannot be suppressed. Finally, we show that CD4(+)CD25(+) T reg cells develop normally in the absence of TGF-beta1 and retain the ability to suppress colitis in vivo. Importantly, the function of TGF-beta1(-/-) T reg cells was abrogated by anti-TGF-beta monoclonal antibody, indicating that functional TGF-beta can be provided by a non-T reg cell source.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Tolerancia Inmunológica/inmunología , Receptores de Interleucina-2/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Citocinas/metabolismo , Ratones , Ratones Noqueados , Receptores de Interleucina-2/inmunología , Bazo/inmunología , Bazo/metabolismo , Células TH1/citología , Células TH1/metabolismo , Timo/inmunología , Timo/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
14.
Int J Nanomedicine ; 16: 4451-4470, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234436

RESUMEN

BACKGROUND: Liver fibrosis is a chronic liver disease with excessive production of extracellular matrix proteins, leading to cirrhosis, hepatocellular carcinoma, and death. PURPOSE: This study aimed at the development of a novel derivative of polyethyleneimine (PEI) that can effectively deliver transforming growth factor ß (TGFß) siRNA and inhibit chemokine receptor 4 (CXCR4) for TGFß silencing and CXCR4 Inhibition, respectively, to treat CCl4-induced liver fibrosis in a mouse model. METHODS: Cyclam-modified PEI (PEI-Cyclam) was synthesized by incorporating cyclam moiety into PEI by nucleophilic substitution reaction. Gel electrophoresis confirmed the PEI-Cyclam polyplex formation and stability against RNAase and serum degradation. Transmission electron microscopy and zeta sizer were employed for the morphology, particle size, and zeta potential, respectively. The gene silencing and CXCR4 targeting abilities of PEI-Cyclam polyplex were evaluated by luciferase and CXCR4 redistribution assays, respectively. The histological and immunohistochemical staining determined the anti-fibrotic activity of PEI-Cyclam polyplex. The TGFß silencing of PEI-Cyclam polyplex was authenticated by Western blotting. RESULTS: The 1H NMR of PEI-Cyclam exhibited successful incorporation of cyclam content onto PEI. The PEI-Cyclam polyplex displayed spherical morphology, positive surface charge, and stability against RNAse and serum degradation. Cyclam modification decreased the cytotoxicity and demonstrated CXCR4 antagonistic and luciferase gene silencing efficiency. PEI-Cyclam/siTGFß polyplexes decreased inflammation, collagen deposition, apoptosis, and cell proliferation, thus ameliorating liver fibrosis. Also, PEI-Cyclam/siTGFß polyplex significantly downregulated α-smooth muscle actin, TGFß, and collagen type III. CONCLUSION: Our findings validate the feasibility of using PEI-Cyclam as a siRNA delivery vector for simultaneous TGFß siRNA delivery and CXCR4 inhibition for the combined anti-fibrotic effects in a setting of CCl4-induced liver fibrosis.


Asunto(s)
Tetracloruro de Carbono/efectos adversos , Compuestos Heterocíclicos/química , Cirrosis Hepática/genética , Polietileneimina/química , ARN Interferente Pequeño/genética , Factor de Crecimiento Transformador beta/genética , Animales , Apoptosis/efectos de los fármacos , Portadores de Fármacos/química , Silenciador del Gen , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Ratones , Tamaño de la Partícula , ARN Interferente Pequeño/química , Receptores CXCR4/genética , Factor de Crecimiento Transformador beta/deficiencia
15.
J Exp Med ; 184(6): 2417-22, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8976197

RESUMEN

Transforming growth factor beta 1 (TGF-beta 1) regulates leukocytes and epithelial cells. To determine whether the pleiotropic effects of TGF-beta 1, a cytokine that is produced by both keratinocytes and Langerhans cells (LC), extend to epidermal leukocytes, we characterized LC (the epidermal contingent of the dendritic cell [DC] lineage) and dendritic epidermal T cells (DETC) in TGF-beta 1 null (TGF-beta 1 -/-) mice. I-A+ LC were not detected in epidermal cell suspensions or epidermal sheets prepared from TGF-beta 1 -/- mice, and epidermal cell suspensions were devoid of allostimulatory activity. In contrast, TCR-gamma delta + DETC were normal in number and appearance in TGF-beta 1 -/- mice and, importantly, DETC represented the only leukocytes in the epidermis. Immunolocalization studies revealed CD11c+ DC in lymph nodes from TGF-beta 1 -/- mice, although gp40+ DC were absent. Treatment of TGF-beta 1 -/- mice with rapamycin abrogated the characteristic inflammatory wasting syndrome and prolonged survival indefinitely, but did not result in population of the epidermis with LC. Thus, the LC abnormality in TGF-beta 1 -/- mice is not a consequence of inflammation in skin or other organs, and LC development is not simply delayed in these animals. We conclude that endogenous TGF-beta 1 is essential for normal murine LC development or epidermal localization.


Asunto(s)
Células de Langerhans/inmunología , Piel/citología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Técnicas de Cocultivo , Femenino , Citometría de Flujo , Inmunosupresores/farmacología , Células de Langerhans/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fenotipo , Polienos/farmacología , Sirolimus , Piel/inmunología , Factor de Crecimiento Transformador beta/genética
16.
J Exp Med ; 199(1): 5-14, 2004 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-14707111

RESUMEN

Elucidation of pathways involved in mouse strain-dependent variation in the hematopoietic stem cell (HSC) compartment may reveal novel mechanisms relevant in vivo. Here, we demonstrate genetically determined variation in the proliferation of lin-Sca1++kit+ (LSK) primitive hematopoietic progenitor cells in response to transforming growth factor-beta (TGF-beta) 2, the dose response of which was biphasic with a stimulatory effect at low concentrations. In contrast, the dose responses of TGF-beta1 or -beta3 were inhibitory and did not show mouse strain-dependent variation. A quantitative trait locus (QTL) for the effect of TGF-beta2 was identified on chromosome 4 overlapping with a QTL regulating the frequency of LSK cells. These overlapping QTL were corroborated by the observation that the frequency of LSK cells is lower in adult Tgfb2+/- mice than in wild-type littermates, indicating that TGF-beta2 is a genetically determined positive regulator LSK number in vivo. Furthermore, adult Tgfb2+/- mice have a defect in competitive repopulation potential that becomes more pronounced upon serial transplantation. In fetal TGF-beta2-deficient HSCs, a defect only appears after serial reconstitution. These data suggest that TGF-beta2 can act cell autonomously and is important for HSCs that have undergone replicative stress. Thus, TGF-beta2 is a novel, genetically determined positive regulator of adult HSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Sitios de Carácter Cuantitativo/genética , Células Madre/citología , Factor de Crecimiento Transformador beta/genética , Animales , Mapeo Cromosómico , Células Madre Hematopoyéticas/fisiología , Hígado/citología , Hígado/embriología , Ratones , Ratones Endogámicos , Ratones Noqueados , Células Madre/fisiología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta2
17.
J Exp Med ; 183(5): 2337-42, 1996 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8642342

RESUMEN

Transforming growth factor beta 1 null mice (TGF-beta 1-/-) suffer from multifocal inflammation and die by 3-4 wk of age. In these mice, levels of nitric oxide (NO) reaction products in serum are elevated approximately fourfold over levels in controls, peaking at 15-17 d of life. Shortterm treatment of TGF-beta 1-/- mice with NG-monomethyl-L-arginine suppressed this elevated production of NO. Expression of inducible NO synthase (iNOS) mRNA and protein is increased in the kidney and heart of TGF-beta 1-/- mice. These findings demonstrate that TGF-beta 1 negatively regulates iNOS expression in vivo, as had been inferred from mechanistic studies on the control of iNOS expression by TGF-beta 1 in vitro.


Asunto(s)
Arginina/análogos & derivados , Expresión Génica/efectos de los fármacos , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/biosíntesis , Factor de Crecimiento Transformador beta/deficiencia , Secuencia de Aminoácidos , Animales , Arginina/farmacología , Northern Blotting , Inducción Enzimática , Genotipo , Inmunohistoquímica , Isoenzimas/análisis , Isoenzimas/antagonistas & inhibidores , Isoenzimas/biosíntesis , Riñón/enzimología , Riñón/patología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Miocardio/enzimología , NG-Nitroarginina Metil Éster , Nitratos/sangre , Óxido Nítrico Sintasa/análisis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitritos/sangre , Fragmentos de Péptidos/inmunología , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Valores de Referencia , Factor de Crecimiento Transformador beta/genética , omega-N-Metilarginina
18.
Mol Cell Biol ; 27(15): 5468-78, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17515606

RESUMEN

The process of implantation, necessary for all viviparous birth, consists of tightly regulated events, including apposition of the blastocyst, attachment to the uterine lumen, and differentiation of the uterine stroma. In rodents and primates the uterine stroma undergoes a process called decidualization. Decidualization, the process by which the uterine endometrial stroma proliferates and differentiates into large epithelioid decidual cells, is critical to the establishment of fetal-maternal communication and the progression of implantation. The role of bone morphogenetic protein 2 (Bmp2) in regulating the transformation of the uterine stroma during embryo implantation in the mouse was investigated by the conditional ablation of Bmp2 in the uterus using the (PR-cre) mouse. Bmp2 gene ablation was confirmed by real-time PCR analysis in the PR-cre; Bmp2fl/fl (termed Bmp2d/d) uterus. While littermate controls average 0.9 litter of 6.2+/-0.7 pups per month, Bmp2d/d females are completely infertile. Analysis of the infertility indicates that whereas embryo attachment is normal in the Bmp2d/d as in control mice, the uterine stroma is incapable of undergoing the decidual reaction to support further embryonic development. Recombinant human BMP2 can partially rescue the decidual response, suggesting that the observed phenotypes are not due to a developmental consequence of Bmp2 ablation. Microarray analysis demonstrates that ablation of Bmp2 leads to specific gene changes, including disruption of the Wnt signaling pathway, Progesterone receptor (PR) signaling, and the induction of prostaglandin synthase 2 (Ptgs2). Taken together, these data demonstrate that Bmp2 is a critical regulator of gene expression and function in the murine uterus.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Decidua/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/deficiencia , Diferenciación Celular , Proliferación Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Decidua/citología , Decidua/patología , Implantación del Embrión , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Infertilidad Femenina/patología , Ratones , Análisis por Micromatrices , Modelos Genéticos , Neovascularización Fisiológica , Ovario/patología , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
19.
Clin Exp Immunol ; 155(1): 65-71, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19076830

RESUMEN

Ulcerative colitis (UC) and Crohn's disease (CD) are considered to be immunologically mediated disorders that share certain features with murine models of colitis. Whether any of these models are physiologically relevant to the human condition remains controversial. The hypothesis is that increased amounts of antibodies neutralizing transforming growth factor (TGF)-beta, interleukin (IL)-2 or IL-10 create a relative immunodeficient state in inflammatory bowel disease (IBD) that predisposes to disease. To evaluate this, serum samples from patients with UC or CD and from normal healthy individuals were studied by enzyme-linked immunosorbent assays. Antibodies recognizing TGF-beta were most prevalent in UC (P<0.01); anti-IL-10 antibodies were elevated in CD (P<0.05), while anti-IL-2 antibodies were the same for all three groups. Importantly, the percentage of IBD patients with at least one of the antibody levels greater than any control value was 30% for UC and 33% for CD. To verify the presence of these antibodies, immobilized TGF-beta was exposed to UC sera and the attached proteins identified by Western blot assay. The proteins proved to be exclusively immunoglobulin (Ig) G. To evaluate the neutralizing activity of these antibodies, cytokine-specific IgG from subjects in each group of patients was incubated with TGF-beta, IL-2 or IL-10 before addition to a bioassay with changes in viability determined by a colorimetric analysis. Antibodies from most individuals in all three groups neutralized the action of each cytokine. This study shows that about one-third of IBD patients may have a relative deficiency of TGF-beta, IL-2 or IL-10 due to an increase in neutralizing antibodies in their sera.


Asunto(s)
Autoanticuerpos/análisis , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Interleucina-10/deficiencia , Interleucina-2/deficiencia , Factor de Crecimiento Transformador beta/deficiencia , Adulto , Análisis de Varianza , Autoanticuerpos/inmunología , Autoanticuerpos/farmacología , Estudios de Casos y Controles , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Inmunoglobulina G/análisis , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Estadísticas no Paramétricas
20.
J Cell Biol ; 162(6): 1135-47, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12963705

RESUMEN

Satellite cells are quiescent muscle stem cells that promote postnatal muscle growth and repair. Here we show that myostatin, a TGF-beta member, signals satellite cell quiescence and also negatively regulates satellite cell self-renewal. BrdU labeling in vivo revealed that, among the Myostatin-deficient satellite cells, higher numbers of satellite cells are activated as compared with wild type. In contrast, addition of Myostatin to myofiber explant cultures inhibits satellite cell activation. Cell cycle analysis confirms that Myostatin up-regulated p21, a Cdk inhibitor, and decreased the levels and activity of Cdk2 protein in satellite cells. Hence, Myostatin negatively regulates the G1 to S progression and thus maintains the quiescent status of satellite cells. Immunohistochemical analysis with CD34 antibodies indicates that there is an increased number of satellite cells per unit length of freshly isolated Mstn-/- muscle fibers. Determination of proliferation rate suggests that this elevation in satellite cell number could be due to increased self-renewal and delayed expression of the differentiation gene (myogenin) in Mstn-/- adult myoblasts. Taken together, these results suggest that Myostatin is a potent negative regulator of satellite cell activation and thus signals the quiescence of satellite cells.


Asunto(s)
Quinasas CDC2-CDC28 , Diferenciación Celular/fisiología , Fibras Musculares Esqueléticas/metabolismo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antígenos CD34/metabolismo , División Celular/fisiología , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Regulación hacia Abajo/fisiología , Retroalimentación Fisiológica/genética , Fase G1/genética , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Miostatina , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/genética , Células Satélite del Músculo Esquelético/citología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda