Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
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
2.
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
3.
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
4.
FEBS J ; 285(1): 101-114, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29117645

RESUMEN

TGFBIp is a constituent of the extracellular matrix in many human tissues including the cornea, where it is one of the most abundant proteins expressed. TGFBIp interacts with Type I, II, IV, VI, and XII collagens as well as several members of the integrin family, suggesting it plays an important role in maintaining structural integrity and possibly corneal transparency as well. Significantly, more than 60 point mutations within the TGFBI gene have been reported to result in aberrant TGFBIp folding and aggregation in the cornea, resulting in severe visual impairment and blindness. Several studies have focused on targeting TGFBIp in the cornea as a therapeutic approach to treat TGFBI-linked corneal dystrophies, but the effect of this approach on corneal homeostasis and matrix integrity remained unknown. In the current study, we evaluated the histological and proteomic profiles of corneas from TGFBI-deficient mice as well as potential redundant functions of the paralogous protein POSTN. The absence of TGFBIp in mouse corneas did not grossly affect the collagen scaffold, and POSTN is unable to compensate for loss of TGFBIp. Proteomic comparison of wild-type and TGFBI-/- mice revealed 11 proteins were differentially regulated, including Type VI and XII collagens. However, as these alterations did not manifest at the macroscopic and behavioral levels, these data support partial or complete TGFBI knockdown as a potential therapy against TGFBI-linked corneal dystrophies. Lastly, in situ hybridization verified TGFBI mRNA in the epithelial cells but not in other cell types, supportive of a therapy directed specifically at this lineage.


Asunto(s)
Córnea/metabolismo , Distrofias Hereditarias de la Córnea/metabolismo , Matriz Extracelular/metabolismo , Proteómica/métodos , Factor de Crecimiento Transformador beta/deficiencia , Anciano , Anciano de 80 o más Años , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Córnea/ultraestructura , Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/terapia , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Factor de Crecimiento Transformador 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.
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
7.
Artículo en Inglés | MEDLINE | ID: mdl-28504090

RESUMEN

Immune responses have a crucial role during the wound healing process in cutaneous leishmaniasis (CL). However, there are several paradoxes in immunity against CL. On the one hand, regulatory cytokines interleukin (IL)-10 and transforming growth factor beta (TGF-ß) increase susceptibility to CL through suppression of several proinflammatory cytokines that require for defense against CL. On the other hand, these cytokines play a pivotal role in the acceleration of wound healing process. This review discusses about the dual role of IL-10 and TGF-ß during the wound healing process and immunity against CL to offer a new insight about wound healing in CL.


Asunto(s)
Interleucina-10/inmunología , Leishmaniasis Cutánea/inmunología , Factor de Crecimiento Transformador beta/inmunología , Cicatrización de Heridas/inmunología , Animales , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Ratones , Células Th17 , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
8.
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
9.
Mech Dev ; 143: 53-61, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28093265

RESUMEN

Gonadal soma-derived factor (gsdf) is reported to be a male initiator in medaka based on loss- and gain- of function via targeted disruption, or transgenic over-expression. However, little is known about how gsdf promotes undifferentiated gonad entry into male pathways or prevents entry into the female pathway. We utilized a visible folliculogenesis system with a reporter cassette of dual-color fluorescence expression to identify difference between oocyte development from wildtype and gsdf deficiency medaka. A red fluorescent protein (RFP) is driven by a major component of the synaptonemal complex (SYCP3) promoter which enables RFP expression solely in oocytes after the onset of meiosis, and a histone 2b-EGFP fused protein (H2BEGFP) under the control of an elongation factor (EF1α) promoter, wildly used as a mitotic reporter of cell cycle. This mitosis-meiosis visible switch revealed that early meiotic oocytes present in gsdf deficiency were more than those in wildtype ovaries, corresponding to the decrease of inhibin expression detected by real-time qPCR analysis, suggesting gsdf is tightly involved in the process of medaka oocyte development at early stage.


Asunto(s)
Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica , Inhibinas/genética , Oocitos/metabolismo , Oryzias/genética , Factor de Crecimiento Transformador beta/genética , Animales , Embrión no Mamífero , Femenino , Proteínas de Peces/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/genética , Histonas/metabolismo , Inhibinas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Meiosis , Mitosis , Oocitos/citología , Oocitos/crecimiento & desarrollo , Oryzias/crecimiento & desarrollo , Oryzias/metabolismo , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Proteína Fluorescente Roja
10.
ACS Nano ; 8(4): 3636-45, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24580381

RESUMEN

Achievement of potent immunoresponses against self/tumor antigens and effective therapeutic outcome against advanced tumors remain major challenges in cancer immunotherapy. The specificity and efficiency of two nanoparticle-based delivery systems, lipid-calcium-phosphate (LCP) nanoparticle (NP) and liposome-protamine-hyaluronic acid (LPH) NP, provide us an opportunity to address both challenges. A mannose-modified LCP NP delivered both tumor antigen (Trp 2 peptide) and adjuvant (CpG oligonucleotide) to the dendritic cells and elicited a potent, systemic immune response regardless of the existence or the stage of tumors in the host. This vaccine was less effective, however, against later stage B16F10 melanoma in a subcutaneous syngeneic model. Mechanistic follow-up studies suggest that elevated levels of immune-suppressive cytokines within the tumor microenvironment, such as TGF-ß, might be responsible. We strategically augment the efficacy of LCP vaccine on an advanced tumor by silencing TGF-ß in tumor cells. The delivery of siRNA using LPH NP resulted in about 50% knockdown of TGF-ß in the late stage tumor microenvironment. TGF-ß down-regulation boosted the vaccine efficacy and inhibited tumor growth by 52% compared with vaccine treatment alone, as a result of increased levels of tumor infiltrating CD8+ T cells and decreased level of regulatory T cells. Combination of systemic induction of antigen-specific immune response with LCP vaccine and targeted modification of tumor microenvironment with LPH NP offers a flexible and powerful platform for both mechanism study and immunotherapeutic strategy development.


Asunto(s)
Melanoma Experimental/prevención & control , Nanopartículas/química , ARN Interferente Pequeño/química , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Microambiente Tumoral/inmunología , Vacunación/métodos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Fosfatos de Calcio/química , Proliferación Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Progresión de la Enfermedad , Regulación hacia Abajo/inmunología , Portadores de Fármacos/química , Femenino , Ácido Hialurónico/química , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Protaminas/química , ARN Interferente Pequeño/genética , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/genética
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.
Curr Opin Immunol ; 23(5): 660-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21839624

RESUMEN

TGF-ß is a cytokine required for the induction and maintenance of transplantation tolerance in animal models. TGF-ß mediates anti-inflammatory effects by acting on many immune cell-types. Central for transplantation tolerance is the role for TGF-ß in the induction of Foxp3 and regulatory capacity in CD4(+) T cells. Recently, however, the general anti-inflammatory role of TGF-ß in CD4(+) T cell polarization was questioned by the discovery that, in the presence of inflammatory cytokines such as IL-6 or IL-1, TGF-ß drives the differentiation of Th17 cells associated with transplant rejection. A better understanding of the factors determining TGF-ß production and activation, Foxp3 induction and Treg stability is vital for the development of tolerogenic strategies in transplantation.


Asunto(s)
Inmunidad Adaptativa , Factores de Transcripción Forkhead/inmunología , Rechazo de Injerto/prevención & control , Trasplante de Órganos , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta , Tolerancia al Trasplante , Animales , Antígenos CD/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/genética , Rechazo de Injerto/inmunología , Humanos , Interleucina-1/inmunología , Interleucina-6/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Linfocitos T Reguladores/citología , Células Th17/citología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/farmacología
13.
Cancer Res ; 69(8): 3415-24, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351843

RESUMEN

Although constitutively activated nuclear factor-kappaB (NF-kappaB), attenuated transforming growth factor beta (TGFbeta) signaling, and TP53 mutations frequently occur in human cancers, how these pathways interact and together contribute to malignancy remains uncertain. Here, we found an association between overexpression of NF-kappaB-related genes, reduced expression of TGFbeta receptor (TbetaR) subunits and downstream targets, and TP53 genotype in head and neck squamous cell carcinoma (HNSCC). In response to recombinant TGFbeta1, both growth inhibition and TGFbeta target gene modulation were attenuated or absent in a panel of human HNSCC lines. However, in HNSCC cells that retained residual TGFbeta signaling, TGFbeta1 inhibited both constitutive and tumor necrosis factor alpha-stimulated NF-kappaB activity. Furthermore, HNSCC lines overexpressing mutant (mt) TP53 and human tumor specimens with positive TP53 nuclear staining exhibited reduced TbetaRII and knocking down mtTP53 induced TbetaRII, increasing TGFbeta downstream gene expression while inhibiting proinflammatory NF-kappaB target gene expression. Transfection of ectopic TbetaRII directly restored TGFbeta signaling while inhibiting inhibitor kappaBalpha degradation and suppressing serine-536 phosphorylation of NF-kappaB p65 and NF-kappaB transcriptional activation, linking these alterations. Finally, experiments with TbetaRII conditional knockout mice show that abrogation of TGFbeta signaling promotes the sustained induction of NF-kappaB and its proinflammatory target genes during HNSCC tumorigenesis and progression. Together, these findings elucidate a regulatory framework in which attenuated TGFbeta signaling promotes NF-kappaB activation and squamous epithelial malignancy in the setting of altered TP53 status.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , FN-kappa B/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoglicanos/biosíntesis , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Transcripción Genética , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
14.
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
15.
J Am Soc Nephrol ; 19(11): 2181-91, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18923055

RESUMEN

Individuals with congenital renal hypoplasia display a defect in the growth of nephrons during development. Many genes that affect the initial induction of nephrons have been identified, but little is known about the regulation of postinductive stages of kidney development. In the absence of the growth factor bone morphogenic protein 7 (BMP7), kidney development arrests after induction of a small number of nephrons. The role of BMP7 after induction, however, has not been fully investigated. Here, we generated a podocyte-specific conditional knockout of BMP7 (Bmp7(flox/flox);Nphs2-Cre(+) [BMP7 CKO]) to study the role of podocyte-derived BMP7 in nephron maturation. By postnatal day 4, 65% of BMP7 CKO mice had hypoplastic kidneys, but glomeruli demonstrated normal patterns of laminin and collagen IV subunit expression. Developing proximal tubules, however, were reduced in number and demonstrated impaired cellular proliferation. We examined signaling pathways downstream of BMP7; the level of cortical phosphorylated Smad1, 5, and 8 was unchanged in BMP CKO kidneys, but phosphorylated p38 mitogen-activated protein kinase was significantly decreased. In addition, beta-catenin was reduced in BMP7 CKO kidneys, and its localization to intracellular vesicles suggested that it had been targeted for degradation. In summary, these results define a BMP7-mediated regulatory axis between glomeruli and proximal tubules during kidney development.


Asunto(s)
Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/metabolismo , Nefronas/crecimiento & desarrollo , Nefronas/metabolismo , Podocitos/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteína Morfogenética Ósea 7 , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular , Colágeno Tipo IV/metabolismo , Humanos , Glomérulos Renales/crecimiento & desarrollo , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Túbulos Renales Proximales/crecimiento & desarrollo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Laminina/metabolismo , Ratones , Ratones Noqueados , Nefronas/patología , Podocitos/patología , Transducción de Señal , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/genética , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Int J Mol Med ; 22(4): 473-80, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18813854

RESUMEN

The transforming growth factor-beta (TGF-beta) has been identified as an important component of wound healing. Recent developments in molecular therapy offer good prospects for the modulation of wound healing, specifically those targeting TGF-beta. The aim of this study was to analyze the effect of TGF-beta targeting on the expression of angiogenic vascular endothelial growth factor (VEGF), a key regulator of angiogenesis and in vitro angiogenic activity in fibroblasts isolated from radiation-induced chronic dermal wounds. The expression of angiogenic VEGF in tissue samples from radiation-induced chronic dermal wounds was investigated by immunohistochemistry and microarray technique. The effect of TGF-beta targeting using antisense oligonucleotides on the expression of VEGF in isolated fibroblasts was analyzed by ELISA and multiplex RT-PCR. Human endothelial cells (ECs) were grown in conditioned medium produced from the treated fibroblasts. EC migration was measured using a modified Boyden chamber; EC tube formation was analyzed under a light microscope. Immunohistochemical investigation and microarray analysis demonstrated a decreased expression of VEGF protein and mRNA in tissue samples from radiation-induced chronic dermal wounds compared to normal human skin. Antisense TGF-beta oligonucleotide treatment significantly up-regulated VEGF secretion in vitro. Addition of conditioned medium from TGF-beta antisense-treated fibroblasts resulted in an increase in EC cell migration and tube formation. In conclusion, our results demonstrate that TGF-beta antisense oligonucleotide technology may be a potential therapeutic option for stimulation of angiogenesis in radiation-induced dermal wounds.


Asunto(s)
Fibroblastos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Piel/patología , Piel/efectos de la radiación , Factor de Crecimiento Transformador beta/deficiencia , Factor A de Crecimiento Endotelial Vascular/genética , Movimiento Celular/efectos de los fármacos , Separación Celular , Medios de Cultivo Condicionados , Citocinas/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Mol Cell Endocrinol ; 294(1-2): 70-80, 2008 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-18790002

RESUMEN

The role transforming growth factor beta (TGFb) isoforms TGFb1, TGFb2 and TGFb3 have in the regulation of embryonic gonadal development was investigated with the use of null-mutant (i.e. knockout) mice for each of the TGFb isoforms. Late embryonic gonadal development was investigated because homozygote TGFb null-mutant mice generally die around birth, with some embryonic loss as well. In the testis, the TGFb1 null-mutant mice had a decrease in the number of germ cells at birth, postnatal day 0 (P0). In the testis, the TGFb2 null-mutant mice had a decrease in the number of seminiferous cords at embryonic day 15 (E15). In the ovary, the TGFb2 null-mutant mice had an increase in the number of germ cells at P0. TGFb isoforms appear to have a role in gonadal development, but interactions between the isoforms is speculated to compensate in the different TGFb isoform null-mutant mice.


Asunto(s)
Desarrollo Embrionario/genética , Mutación/genética , Ovario/embriología , Ovario/metabolismo , Testículo/embriología , Testículo/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Animales , Antígenos Nucleares/metabolismo , Apoptosis , Recuento de Células , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Genotipo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/citología , Fenotipo , Ratas , Túbulos Seminíferos/citología , Túbulos Seminíferos/embriología , Túbulos Seminíferos/metabolismo , Razón de Masculinidad , Testículo/citología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/deficiencia , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/deficiencia , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo
18.
J Am Soc Nephrol ; 19(11): 2098-107, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18632843

RESUMEN

In diabetic nephropathy, connective tissue growth factor (CTGF) is upregulated and bone morphogenetic protein 7 (BMP-7) is downregulated. CTGF is known to inhibit BMP-4, but similar cross-talk between BMP-7 and CTGF has not been studied. In this study, it was hypothesized that CTGF acts as an inhibitor of BMP-7 signaling activity in diabetic nephropathy. Compared with diabetic wild-type CTGF(+/+) mice, diabetic CTGF(+/-) mice had approximately 50% lower CTGF mRNA and protein, less severe albuminuria, no thickening of the glomerular basement membrane, and preserved matrix metalloproteinase (MMP) activity. Although the amount of BMP-7 mRNA was similar in the kidneys of diabetic CTGF(+/+) and CTGF(+/-) mice, phosphorylation of the BMP signal transduction protein Smad1/5 and expression of the BMP target gene Id1 were lower in diabetic CTGF(+/+) mice. Moreover, renal Id1 mRNA expression correlated with albuminuria (R = -0.86) and MMP activity (R = 0.76). In normoglycemic mice, intraperitoneal injection of CTGF led to a decrease of pSmad1/5 in the renal cortex. In cultured renal glomerular and tubulointerstitial cells, CTGF diminished BMP-7 signaling activity, evidenced by lower levels of pSmad1/5, Id1 mRNA, and BMP-responsive element-luciferase activity. Co-immunoprecipitation, solid-phase binding assay, and surface plasmon resonance analysis showed that CTGF binds BMP-7 with high affinity (Kd approximately 14 nM). In conclusion, upregulation of CTGF inhibits BMP-7 signal transduction in the diabetic kidney and contributes to altered gene transcription, reduced MMP activity, glomerular basement membrane thickening, and albuminuria, all of which are hallmarks of diabetic nephropathy.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Nefropatías Diabéticas/fisiopatología , Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Factor de Crecimiento del Tejido Conjuntivo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Femenino , Expresión Génica , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/farmacología , Proteína 1 Inhibidora de la Diferenciación/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
19.
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
20.
Eur J Hum Genet ; 16(1): 36-44, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17878916

RESUMEN

Urogenital birth defects are one of the common phenotypes observed in hereditary human disorders. In particular, limb malformations are often associated with urogenital developmental abnormalities, as the case for Hand-foot-genital syndrome displaying similar hypoplasia/agenesis of limbs and external genitalia. Split-hand/split-foot malformation (SHFM) is a syndromic limb disorder affecting the central rays of the autopod with median clefts of the hands and feet, missing central fingers and often fusion of the remaining ones. SHFM type 1 (SHFM1) is linked to genomic deletions or rearrangements, which includes the distal-less-related homeogenes DLX5 and DLX6 as well as DSS1. SHFM type 4 (SHFM4) is associated with mutations in p63, which encodes a p53-related transcription factor. To understand that SHFM is associated with urogenital birth defects, we performed gene expression analysis and gene knockout mouse model analyses. We show here that Dlx5, Dlx6, p63 and Bmp7, one of the p63 downstream candidate genes, are all expressed in the developing urethral plate (UP) and that targeted inactivation of these genes in the mouse results in UP defects leading to abnormal urethra formation. These results suggested that different set of transcription factors and growth factor genes play similar developmental functions during embryonic urethra formation. Human SHFM syndromes display multiple phenotypes with variations in addition to split hand foot limb phenotype. These results suggest that different genes associated with human SHFM could also be involved in the aetiogenesis of hypospadias pointing toward a common molecular origin of these congenital malformations.


Asunto(s)
Deformidades Congénitas de las Extremidades/genética , Uretra/anomalías , Animales , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Modelos Animales de Enfermedad , Deformidades Congénitas del Pie/embriología , Deformidades Congénitas del Pie/genética , Regulación del Desarrollo de la Expresión Génica , Genitales/embriología , Deformidades Congénitas de la Mano/embriología , Deformidades Congénitas de la Mano/genética , Proteínas de Homeodominio/genética , Humanos , Deformidades Congénitas de las Extremidades/clasificación , Deformidades Congénitas de las Extremidades/embriología , Ratones , Ratones Noqueados , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Síndrome , Transactivadores/deficiencia , Transactivadores/genética , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética , Uretra/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...