Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Intervalo de año de publicación
1.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33228049

RESUMEN

The inhalation of metal (including lead) nanoparticles poses a real health issue to people and animals living in polluted and/or industrial areas. In this study, we exposed mice to lead(II) nitrate nanoparticles [Pb(NO3)2 NPs], which represent a highly soluble form of lead, by inhalation. We aimed to uncover the effects of their exposure on individual target organs and to reveal potential variability in the lead clearance. We examined (i) lead biodistribution in target organs using laser ablation and inductively coupled plasma mass spectrometry (LA-ICP-MS) and atomic absorption spectrometry (AAS), (ii) lead effect on histopathological changes and immune cells response in secondary target organs and (iii) the clearance ability of target organs. In the lungs and liver, Pb(NO3)2 NP inhalation induced serious structural changes and their damage was present even after a 5-week clearance period despite the lead having been almost completely eliminated from the tissues. The numbers of macrophages significantly decreased after 11-week Pb(NO3)2 NP inhalation; conversely, abundance of alpha-smooth muscle actin (α-SMA)-positive cells, which are responsible for augmented collagen production, increased in both tissues. Moreover, the expression of nuclear factor κB (NF-κB) and selected cytokines, such as tumor necrosis factor alpha (TNFα), transforming growth factor beta 1 (TGFß1), interleukin 6(IL-6), IL-1α and IL-1ß , displayed a tissue-specific response to lead exposure. In summary, diminished inflammatory response in tissues after Pb(NO3)2 NPs inhalation was associated with prolonged negative effect of lead on tissues, as demonstrated by sustained pathological changes in target organs, even after long clearance period.


Asunto(s)
Contaminantes Atmosféricos/farmacocinética , Plomo/farmacocinética , Pulmón/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Nitratos/farmacocinética , Actinas/agonistas , Actinas/genética , Actinas/inmunología , Administración por Inhalación , Contaminantes Atmosféricos/toxicidad , Animales , Disponibilidad Biológica , Femenino , Expresión Génica , Semivida , Exposición por Inhalación/análisis , Interleucina-1alfa/agonistas , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1beta/agonistas , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/agonistas , Interleucina-6/genética , Interleucina-6/inmunología , Plomo/toxicidad , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Nanopartículas del Metal/administración & dosificación , Ratones , Ratones Endogámicos ICR , FN-kappa B/agonistas , FN-kappa B/genética , FN-kappa B/inmunología , Nitratos/toxicidad , Espectrofotometría Atómica , Distribución Tisular , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Factor de Necrosis Tumoral alfa/agonistas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
2.
Nutr Neurosci ; 21(3): 210-218, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27996890

RESUMEN

OBJECTIVES: It has been shown that calcitriol and all-trans retinoic acid (ATRA) have modulatory effects on the immune system. The present study investigates the synergistic effects of combination treatment of calcitriol and ATRA in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). METHODS: The mice were allocated to four preventive groups, each consisting of eight animals, ATRA (250 µg/mouse), calcitriol (100 ng/mouse), combination of ATRA and calcitriol (125  µg/mouse and 50 ng/mouse) and vehicle groups. EAE was induced by MOG35-55 peptide in female C57BL/6 mice. Treatments were initiated at day 1 before immunization and continued every other day throughout the study until the day 21 post-immunization. Splenocytes were isolated from EAE-induced mice and the expression of retinoic acid receptor-related orphan receptor gamma t (ROR-γt), Interleukin-17 (IL-17), transforming growth factor beta (TGF-ß), and forkhead box P3 (FOXP3) genes was measured using real-time polymerase chain reaction. RESULTS: The expression of FOXP3 and TGF-ß genes in the splenocytes of combination-treated and calcitriol alone-treated mice was significantly increased compared to vehicle group (P < 0.05). The expression of ROR-γt and IL-17 genes in the splenocytes of ATRA, calcitriol and combination- treated mice was significantly reduced compared to those of vehicle- treated mice (P < 0.05). The relative expression level of ROR-γt was significantly (P < 0.05) lower in the combination group than in the mice treated by ATRA or calcitriol alone. DISCUSSION: This study demonstrated that treatment with combination of calcitriol and ATRA can be considered as a new strategy for MS prevention and treatment.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antiinflamatorios/uso terapéutico , Calcitriol/uso terapéutico , Encefalomielitis Autoinmune Experimental/prevención & control , Regulación de la Expresión Génica/efectos de los fármacos , Bazo/efectos de los fármacos , Tretinoina/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios no Esteroideos/administración & dosificación , Calcitriol/administración & dosificación , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Factores de Transcripción Forkhead/agonistas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Inyecciones Intraperitoneales , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Reproducibilidad de los Resultados , Bazo/inmunología , Bazo/metabolismo , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Tretinoina/administración & dosificación
3.
São Paulo; s.n; s.n; 2018. 93 p. tab, ilus, graf.
Tesis en Portugués | LILACS | ID: biblio-998850

RESUMEN

O fator de crescimento transformante beta tipo 1, TGF-ß1, é uma proteína extracelular homodimérica secretada por vários tipos celulares, que pode ter ação parácrina ou endócrina. Essa proteína está envolvida em processos celulares de diferenciação, proliferação, mobilidade e formação de matriz extracelular. Além disso, é parte importante dos processos de regeneração tecidual, atuando, de maneira decisiva, no reparo, atraindo macrófagos e fibroblastos para o local da injúria e estimulando a angiogênese. Assim, considerando o papel desse peptídeo no processo regenerativo, o uso de TGF-ß1 como proteína terapêutica na área de Bioengenharia Tecidual é bastante promissor. Apesar disso, a venda dessa proteína, para fins terapêuticos, é inexistente no mercado e a proteína recombinante vendida, que só pode ser utilizada em pesquisas científicas, não é produzida nacionalmente e chega a custar R$200.000,00/mg. Nesse contexto, o objetivo do presente trabalho é desenvolver uma metodologia de produção do fator recombinante TGF-ß1 em células de ovário de hamster chinês (CHO), visando à obtenção de níveis altos de rendimento, e, futuramente, a transferência da tecnologia de produção para a iniciativa privada, tornando possível seu uso na Medicina Regenerativa, sozinho ou em combinação com outros fatores de crescimento. O cDNA de TGF-ß1 foi amplificado a partir de um banco de cDNA humano e clonado no vetor proprietário pNU1 de expressão de mamífero. A construção pNU1/TGF-ß1 foi utilizada para transfectar estavelmente células CHO DG44 e uma estratégia de co-amplificação foi utilizada para selecionar células transfectantes com maior número de cópias da sequência correspondente a TGF-ß1. Estas culturas foram submetidas ao processo de amplificação gênica com concentrações crescentes de metotrexato. Ensaios de Western Blot e ELISA foram realizados utilizando-se o meio condicionado pelas populações selecionadas e por clones superprodutores. Entre os 41clones obtidos, cinco apresentaram maiores níveis de produção de TGF-ß1, entre 1.000 e 2.000 ng/mL. Estes clones foram selecionados para a realização de testes de atividade in vitro utilizando-se células A549, que permitem avaliar a transição epitélio-mesênquima. Um ensaio de cicatrização de feridas em peles do dorso de camundongos foi padronizado e utilizado para avaliar a atividade in vivo do clone que apresentou melhor resultado in vitro. A proteína TGF-ß1 foi parcialmente purificada por HPLC em uma coluna de afinidade. Portanto, a proteína TGF-ß1 humana recombinante foi produzida, apresentando atividade biológica in vitro e in vivo, sendo capaz de reparar eficientemente feridas cutâneas. Essa iniciativa pode oferecer aos pacientes uma alternativa para o tratamento de lesões teciduais, acelerando a cicatrização de feridas e o reparo de tecidos


The transforming growth factor beta 1, TGF-ß1, is a homodimeric extracellular protein secreted by several cell types, which may have paracrine or endocrine action. This protein is involved in cellular processes of differentiation, proliferation, mobility and formation of extracellular matrix. In addition, it is an important part of the tissue regeneration processes, acting decisively on repair, attracting macrophages and fibroblasts to the site of injury and stimulating angiogenesis. Therefore, considering the role of this peptide in the regenerative process and the use of TGF-ß1 as a therapeutic protein in the field of Tissue Bioengineering is very promising. Despite this, the sale of this protein for therapeutic purposes is nonexistent in the market and the recombinant protein available in the market, which can only be used in scientific research, is not produced nationally and the costs are in the order of R$ 200,000.00/mg. In this context, the objective of the present work is to develop a methodology for the production of the TGF-ß1 recombinant factor in Chinese hamster ovary (CHO) cells, aiming at obtaining high yields, and, in the future, transfering the production technology to the private initiative, allowing its use in Regenerative Medicine, alone or in combination with other growth factors. The TGF-ß1 cDNA was amplified from a human cDNA library and cloned into the proprietary pNU1 mammalian expression vector. The pNU1/TGF-ß1 construct was used to stably transfect CHO DG44 cells, and a co-amplification strategy was used to select transfectant cells with the largest number of gene copies. These cultures were subjected to the process of gene amplification with methotrexate. Western Blot and ELISA were used to assay the conditioned medium obtained from the selected cell populations and from overproducing cell clones. Among the 41 clones obtained, five presented higher levels of TGF-ß1 production, between 1,000 and 2,000 ng/mL. These clones were selected for in vitro activity testing using A549 cells to evaluate the epithelial-mesenchymal transition. Awound healing assay on mouse dorsal skin was standardized and used to evaluate the in vivo activity of the cell clone which displayed the highest result in vitro. The TGF-ß1 protein was partially purified by HPLC on an affinity column. Therefore, the recombinant human TGF-ß1 protein was produced and shown to display biological activity both in vitro and in vivo, being able to eficiently repair cutaneous wounds. This initiative may provide patients with an alternative treatment for tissue damage, accelerating wound healing and tissue repair


Asunto(s)
Animales , Ratones , Células CHO/citología , Medicina Regenerativa/clasificación , Factor de Crecimiento Transformador beta1/agonistas , Mamíferos , Técnicas In Vitro , Ensayo de Inmunoadsorción Enzimática , Western Blotting , Cromatografía Líquida de Alta Presión/instrumentación
4.
DNA Cell Biol ; 36(7): 603-610, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28467108

RESUMEN

Gremlin1, the antagonist of bone morphogenetic protein-7 and one of the target genes of transforming growth factor (TGF)-ß signal pathway, plays an important role in embryonic development and its expression decreases along with aging. To explore the expression of gremlin1 in liver fibrosis and the causal link between gremlin1 and hepatic stellate cell (HSC) activation, we detected the expression of gremlin1 in mice with hepatic fibrosis induced by porcine serum using real time quantitative PCR (RT-qPCR) and immunohistochemical staining. The hepatic fibrosis mice were evaluated by the external feature of the liver, histology, hepatic function, collagen deposition, and the expression of fibrosis-related genes (genes COLIα2 and COLIVα2) in the liver. In the HSC-T6, western blotting was used to analyze the expression of α-smooth muscle actin (α-SMA), COL1α, and TGF-ß1 in conditions of overexpression of gremlin1 or gremlin1 being knocked down by specific siRNA, respectively. The results showed that the mRNA expression of the gremlin1 gene was significantly increased consistent with increased expression of COLIα2 and COLIVα2 in the liver tissue of the hepatic fibrosis mice. Increased expression of gremlin1 coincided with the same area of the collagen deposition. Furthermore, the results also showed that the expression of α-SMA, COLIα1, and TGF-ß1 was consistent with the expression of gremlin1 not only in the HSC-T6 overexpressing gremlin1 but also in the HSC-T6 that gremlin1 is knocked down by specific siRNA. The findings suggest that gremlin1 might play an important role in the progression of hepatic fibrosis and that it modulates HSC activation.


Asunto(s)
Actinas/genética , Colágeno Tipo I/genética , Regulación Neoplásica de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Cirrosis Hepática/genética , Factor de Crecimiento Transformador beta1/genética , Actinas/agonistas , Actinas/metabolismo , Animales , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Colágeno Tipo I/agonistas , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Estrelladas Hepáticas/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos BALB C , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Suero/química , Transducción de Señal , Porcinos/sangre , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/metabolismo
5.
J Enzyme Inhib Med Chem ; 32(1): 624-631, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28260395

RESUMEN

Fibrotic diseases have become a major cause of death in the developed world. AdipoR1 agonists are potent inhibitors of fibrotic responses. Here, we focused on the in silico identification of novel AdipoR1 peptide agonists. A homology model was constructed to predict the 3D structure of AdipoR1. By docking to known active peptides, the putative active site of the model was further explored. A virtual screening study was then carried out with a set of manually designed peptides using molecular docking. Peptides with high docking scores were then evaluated for their anti-fibrotic properties. The data indicated that the novel peptide Pep70 significantly inhibited the proliferation of hepatic stellate cells (HSC) and NIH-3T3 cells (18.33% and 27.80%) and resulted in favouring cell-cycle arrest through increasing the accumulation of cells in the G0/G1 phase by 17.08% and 15.86%, thereby reducing the cell population in the G2/M phase by 11.25% and 15.95%, respectively. Additionally, Pep70 exhibited the most marked suppression on the expression of α-smooth muscle actin (α-SMA), collagen type I alpha1 (COL1A1) and TGF-ß1. Therefore, the peptide Pep70 was ultimately identified as an inhibitor of fibrotic responses and as a potential AdipoR1 agonist.


Asunto(s)
Oligopéptidos/química , Péptidos/farmacología , Receptores de Adiponectina/agonistas , Secuencia de Aminoácidos , Animales , Sitios de Unión/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Células 3T3 NIH , Oligopéptidos/genética , Péptidos/síntesis química , Péptidos/química , Ratas , Homología de Secuencia de Aminoácido , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/metabolismo
6.
Nature ; 542(7639): 55-59, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28117447

RESUMEN

Integrins are adhesion receptors that transmit force across the plasma membrane between extracellular ligands and the actin cytoskeleton. In activation of the transforming growth factor-ß1 precursor (pro-TGF-ß1), integrins bind to the prodomain, apply force, and release the TGF-ß growth factor. However, we know little about how integrins bind macromolecular ligands in the extracellular matrix or transmit force to them. Here we show how integrin αVß6 binds pro-TGF-ß1 in an orientation biologically relevant for force-dependent release of TGF-ß from latency. The conformation of the prodomain integrin-binding motif differs in the presence and absence of integrin binding; differences extend well outside the interface and illustrate how integrins can remodel extracellular matrix. Remodelled residues outside the interface stabilize the integrin-bound conformation, adopt a conformation similar to earlier-evolving family members, and show how macromolecular components outside the binding motif contribute to integrin recognition. Regions in and outside the highly interdigitated interface stabilize a specific integrin/pro-TGF-ß orientation that defines the pathway through these macromolecules which actin-cytoskeleton-generated tensile force takes when applied through the integrin ß-subunit. Simulations of force-dependent activation of TGF-ß demonstrate evolutionary specializations for force application through the TGF-ß prodomain and through the ß- and not α-subunit of the integrin.


Asunto(s)
Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Integrinas/química , Integrinas/metabolismo , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Evolución Molecular , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Factor de Crecimiento Transformador beta1/metabolismo
7.
Stem Cell Res Ther ; 7(1): 175, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27906099

RESUMEN

BACKGROUND: Epithelial to mesenchymal transition, especially to myofibroblasts, plays an important role in wound healing, fibrosis, and carcinogenesis. Epidermal stem cells (EpSCs) are responsible for epidermal renewal and wound re-epithelialization. However, it remains unclear whether and how EpSCs transdifferentiate into myofibroblasts or myofibroblast-like cells (MFLCs). Here, we provide the first evidence showing that P311 induces EpSC to MFLC transdifferentiation (EpMyT) via TGFß1/Smad signaling. METHODS: Wound healing and mesenchymal features were observed in the P311 KO and P311 WT mouse model of superficial second-degree burns. After the primary human or mouse EpSCs were forced to highly express P311 using an adenoviral vector, EpMyT was observed by immunofluorescence, real-time PCR, and western blot. The activity of TGFß1 and Smad2/3 in EpSCs with different P311 levels was observed by western blot. The TßRI/II inhibitor LY2109761 and Smad3 siRNA were applied to block the EpMyT in P311-overexpressing EpSCs and exogenous TGFß1 was to restore the EpMyT in P311 KO EpSCs. Furthermore, the mechanism of P311 regulating TGFß1 was investigated by bisulfite sequencing PCR, luciferase activity assay, and real-time PCR. RESULTS: P311 KO mouse wounds showed delayed re-epithelialization and reduced mesenchymal features. The human or mouse EpSCs with overexpressed P311 exhibited fusiform morphological changes, upregulated expression of myofibroblast markers (α-SMA and vimentin), and downregulated expression of EpSC markers (ß1-integrin and E-cadherin). P311-expressing EpSCs showed decreased TGFß1 mRNA and increased TGFß1 protein, TßRI/II mRNA, and activated Smad2/3. Moreover, LY2109761 and Smad3 siRNA reversed P311-induced EpMyT. Under the stimulation of exogenous TGFß1, the phosphorylation of Smad2 and Smad3 in P311 KO EpSCs was significantly lower than that in P311 WT EpSCs and the EpMyT in P311 KO EpSCs was restored. Furthermore, P311 enhanced the methylation of TGFß1 promoter and increased activities of TGFß1 5'/3' untranslated regions (UTRs) to stimulate TGFß1 expression. P311+α-SMA+ cells and P311+vimentin+ cells were observed in the epidermis of human burn wounds. Also, P311 was upregulated by IL-1ß, IL-6, TNFα, and hypoxia. CONCLUSIONS: P311 is a novel TGFß1/Smad signaling-mediated regulator of transdifferentiation in EpSCs during cutaneous wound healing. Furthermore, P311 might stimulate TGFß1 expression by promoting TGFß1 promoter methylation and by activating the TGFß1 5'/3' UTR.


Asunto(s)
Quemaduras/genética , Miofibroblastos/metabolismo , Proteínas del Tejido Nervioso/genética , Células Madre/metabolismo , Factor de Crecimiento Transformador beta1/genética , Cicatrización de Heridas/genética , Heridas no Penetrantes/genética , Actinas/genética , Actinas/metabolismo , Animales , Quemaduras/metabolismo , Quemaduras/patología , Cadherinas/genética , Cadherinas/metabolismo , Transdiferenciación Celular , Metilación de ADN , Epidermis/lesiones , Epidermis/metabolismo , Epidermis/patología , Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos/patología , Proteínas del Tejido Nervioso/deficiencia , Cultivo Primario de Células , Regiones Promotoras Genéticas , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Células Madre/patología , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/genética , Vimentina/metabolismo , Heridas no Penetrantes/metabolismo , Heridas no Penetrantes/patología
8.
J Cell Biochem ; 117(4): 938-45, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26369756

RESUMEN

The Wnt ligands are a family of secreted signaling proteins which play key roles in a number of cellular processes under physiological and pathological conditions. Wnts bind to their membrane receptors and initiate a signaling cascade which leads to the nuclear localization and transcriptional activity of ß-catenin. The development of purified recombinant Wnt ligands has greatly aided in our understanding of Wnt signaling and its functions in development and disease. In the current study, we identified non-Wnt related signaling activities which were present in commercially available preparations of recombinant Wnt3a. Specifically, we found that treatment of cultured fibroblasts with recombinant Wnt3a induced immediate activation of TGF-ß and BMP signaling and this activity appeared to be independent of the Wnt ligand itself. Therefore, while purified recombinant Wnt ligands continue to be a useful tool for studying this signaling pathway, one must exercise a degree of caution when analyzing the results of experiments that utilize purified recombinant Wnt ligands.


Asunto(s)
Artefactos , Biotecnología/normas , Factor de Crecimiento Transformador beta1/agonistas , Proteína Wnt3A/farmacología , Animales , Anticuerpos Neutralizantes/farmacología , Biotecnología/métodos , Proteína Morfogenética Ósea 7/antagonistas & inhibidores , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Proteínas/genética , Proteínas/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Control de Calidad , Quinolinas/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
9.
Free Radic Biol Med ; 80: 1-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514442

RESUMEN

Postnatal alveolarization is regulated by a number of growth factors, including insulin-like growth factor-I (IGF-I) acting through the insulin-like growth factor receptor-1 (IGF-R1). Exposure of the neonatal rat lung to 60% O2 for 14 days results in impairments of lung cell proliferation, secondary crest formation, and alveologenesis. This lung injury is mediated by peroxynitrite and is prevented by treatment with a peroxynitrite decomposition catalyst. We hypothesized that one of the mechanisms by which peroxynitrite induces lung injury in 60% O2 is through nitration and inactivation of critical growth factors or their receptors. Increased nitration of both IGF-I and IGF-R1 was evident in 60% O2-exposed lungs, which was reversible by concurrent treatment with a peroxynitrite decomposition catalyst. Increased nitration of the IGF-R1 was associated with its reduced activation, as assessed by IGF-R1 phosphotyrosine content. IGF-I displacement binding plots were conducted in vitro using rat fetal lung distal epithelial cells which respond to IGF-I by an increase in DNA synthesis. When IGF-I was nitrated to a degree similar to that observed in vivo there was minimal, if any, effect on IGF-I displacement binding. In contrast, nitrating cell IGF-R1 to a similar degree to that observed in vivo completely prevented specific binding of IGF-I to the IGF-R1, and attenuated an IGF-I-mediated increase in DNA synthesis. Additionally, we hypothesized that peroxynitrite also impairs alveologenesis by being an upstream regulator of the growth inhibitor, TGFß1. That 60% O2-induced impairment of alveologenesis was mediated in part by TGFß1 was confirmed by demonstrating an improvement in secondary crest formation when 60% O2-exposed pups received concurrent treatment with the TGFß1 activin receptor-like kinase, SB 431542. That the increased TGFß1 content in lungs of pups exposed to 60% O2 was regulated by peroxynitrite was confirmed by its attenuation by concurrent treatment with a peroxynitrite decomposition catalyst. We conclude that peroxynitrite contributes to the impaired alveologenesis observed following the exposure of neonatal rats to 60% O2 both by preventing binding of IGF-I to the IGF-R1, secondary to nitration of the IGF-R1, and by causing an up-regulation of the growth inhibitor, TGFß1.


Asunto(s)
Células Epiteliales/metabolismo , Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Alveolos Pulmonares/metabolismo , Receptores de Somatomedina/metabolismo , Factor de Crecimiento Transformador beta1/agonistas , Animales , Animales Recién Nacidos , Benzamidas/farmacología , ADN/biosíntesis , Dioxoles/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Regulación de la Expresión Génica , Hiperoxia/complicaciones , Hiperoxia/tratamiento farmacológico , Hiperoxia/patología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Metaloporfirinas/farmacología , Nitratos/metabolismo , Ácido Peroxinitroso/antagonistas & inhibidores , Ácido Peroxinitroso/metabolismo , Cultivo Primario de Células , Unión Proteica , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley , Receptores de Somatomedina/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
10.
PLoS One ; 8(8): e73399, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24015303

RESUMEN

BACKGROUND: Pulmonary arterial hypertension is characterized by increased thickness of pulmonary vessel walls due to both increased proliferation of pulmonary arterial smooth muscle cell (PASMC) and deposition of extracellular matrix. In patients suffering from pulmonary arterial hypertension, endothelin-1 (ET-1) synthesis is up-regulated and may increase PASMC activity and vessel wall remodeling through transforming growth factor beta-1 (TGF-ß1) and connective tissue growth factor. OBJECTIVE: To assess the signaling pathway leading to ET-1 induced proliferation and extracellular matrix deposition by human PASMC. METHODS: PASMC were serum starved for 24 hours before stimulation with either ET-1 and/or TGF-ß1. ET-1 was inhibited by Bosentan, ERK1/2 mitogen activated protein kinase (MAPK) was inhibited by U0126 and p38 MAPK was inhibited by SB203580. RESULTS: ET-1 increased PASMC proliferation when combined with serum. This effect involved the mitogen activated protein kinases (MAPK) ERK1/2 MAPK and was abrogated by Bosentan which caused a G1- arrest through activation of p27((Kip)). Regarding the contribution of extracellular matrix deposition in vessel wall remodeling, TGF-ß1 increased the deposition of collagen type-I and fibronectin, which was further increased when ET-1 was added mainly through ERK1/2 MAPK. In contrast, collagen type-IV was not affected by ET-1. Bosentan dose-dependently reduced the stimulatory effect of ET-1 on collagen type-I and fibronectin, but had no effect on TGF-ß1. CONCLUSION AND CLINICAL RELEVANCE: ET-1 alone does not induce PASMC proliferation and extracellular matrix deposition. However, ET-1 significantly up-regulates serum induced proliferation and TGF-ß1 induced extracellular matrix deposition, specifically of collagen type-I and fibronectin. The synergistic effects of ET-1 on serum and TGF-ß1 involve ERK1/2 MAPK and may thus present a novel mode of action in the pathogenesis of pulmonary arterial hypertension.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Endotelina-1/metabolismo , Hipertensión Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Línea Celular , Colágeno Tipo I/biosíntesis , Células Endoteliales/patología , Endotelina-1/agonistas , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Fibronectinas/biosíntesis , Humanos , Hipertensión Pulmonar/patología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Crecimiento Transformador beta1/agonistas
11.
PLoS One ; 8(7): e70479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936211

RESUMEN

Radiation therapy in the treatment of cancer is dose limited by radiation injury in normal tissues such as the intestine and the heart. To identify the mechanistic involvement of transforming growth factor-beta 1 (TGF-ß1) in intestinal and cardiac radiation injury, we studied the influence of pharmacological induction of TGF-ß1 with xaliproden (SR 57746A) in rat models of radiation enteropathy and radiation-induced heart disease (RIHD). Because it was uncertain to what extent TGF-ß induction may enhance radiation injury in heart and intestine, animals were exposed to irradiation schedules that cause mild to moderate (acute) radiation injury. In the radiation enteropathy model, male Sprague-Dawley rats received local irradiation of a 4-cm loop of rat ileum with 7 once-daily fractions of 5.6 Gy, and intestinal injury was assessed at 2 weeks and 12 weeks after irradiation. In the RIHD model, male Sprague-Dawley rats received local heart irradiation with a single dose of 18 Gy and were followed for 6 months after irradiation. Rats were treated orally with xaliproden starting 3 days before irradiation until the end of the experiments. Treatment with xaliproden increased circulating TGF-ß1 levels by 300% and significantly induced expression of TGF-ß1 and TGF-ß1 target genes in the irradiated intestine and heart. Various radiation-induced structural changes in the intestine at 2 and 12 weeks were significantly enhanced with TGF-ß1 induction. Similarly, in the RIHD model induction of TGF-ß1 augmented radiation-induced changes in cardiac function and myocardial fibrosis. These results lend further support for the direct involvement of TGF-ß1 in biological mechanisms of radiation-induced adverse remodeling in the intestine and the heart.


Asunto(s)
Corazón/efectos de los fármacos , Intestinos/efectos de los fármacos , Naftalenos/efectos adversos , Piridinas/efectos adversos , Traumatismos Experimentales por Radiación/inducido químicamente , Factor de Crecimiento Transformador beta1/agonistas , Animales , Biomarcadores/metabolismo , Rayos gamma , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Corazón/fisiopatología , Corazón/efectos de la radiación , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/efectos de la radiación , Masculino , Dosis de Radiación , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/sangre
12.
Biol Reprod ; 89(4): 84, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23926286

RESUMEN

Transforming growth factor-beta (TGF-B) plays an important role in embryo implantation; however, TGF-B requires liberation from its inactive latent forms (i.e., large latent TGF-B complex [LLC] and small latent TGF-B complex [SLC]) to its biologically active (i.e., monomer or dimer) forms in order to act on its receptors (TGF-BRs), which in turn activate SMAD2/3. Activation of TGF-B1 from its latent complexes in the uterus is not yet deciphered. We investigated uterine latent TGF-B1 complex and its biologically active form during implantation, decidualization, and delayed implantation. Our study, utilizing nonreducing SDS-PAGE followed by Western blotting and immunoblotting with TGF-B1, LTBP1, and latency-associated peptide, showed the presence of LLC and SLC in the uterine extracellular matrix and plasma membranous protein fraction during stages of the implantation period. A biologically active form of TGF-B1 (~17-kDa monomer) was highly elevated in the uterine plasma membranous compartment at the peri-implantation stage (implantation and nonimplantation sites). Administration of hydroxychloroquine (an inhibitor of pro-TGF-B processing) at the preimplantation stage was able to block the liberation of biologically active TGF-B1 from its latent complex at the postimplantation stage; as a consequence, the number of implantation sites was reduced at Day 5 (1000 h), as was the number of fetuses at Day 13. The inhibition of TGF-B1 showed reduced levels of phosphorylated SMAD3. Further, the delayed-implantation mouse model showed progesterone and estradiol coordination to release the active TGF-B1 form from its latent complex in the receptive endometrium. This study demonstrates the importance of liberation of biologically active TGF-B1 during the implantation period and its regulation by estradiol.


Asunto(s)
Implantación del Embrión , Endometrio/metabolismo , Estradiol/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Crecimiento Transformador beta1/agonistas , Animales , Decidua/metabolismo , Modelos Animales de Enfermedad , Implantación Tardía del Embrión , Femenino , Infertilidad Femenina/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones , Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Placentación , Embarazo , Progesterona/metabolismo , Precursores de Proteínas/metabolismo , Señales de Clasificación de Proteína , Proteína smad3/agonistas , Proteína smad3/antagonistas & inhibidores , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo
13.
Neurosci Bull ; 24(3): 155-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18500388

RESUMEN

OBJECTIVE: To examine the effect of neuropeptide Y (NPY) on TGF-beta1 production in RAW264.7 macrophages. METHODS: Enzyme linked immunosorbent assay (ELISA) was used to detect TGF-beta1 production. Cell counting kit 8 (CCK-8) was used to assay the viability of RAW264.7 cells. Western blot was used to detect the phosphorylation of PI3K p85. RESULTS: NPY treatment could promote TGF-beta1 production and rapid phosphorylation of PI3K p85 in RAW264.7 cells via Y1 receptor. The elevated TGF-beta1 production induced by NPY could be abolished by wortmannin pretreatment. CONCLUSION: NPY may elicit TGF-beta1 production in RAW264.7 cells via Y1 receptor, and the activated PI3K pathway may account for this effect.


Asunto(s)
Macrófagos/metabolismo , Neuropéptido Y/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Androstadienos/farmacología , Animales , Western Blotting , Recuento de Células , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Ensayo de Inmunoadsorción Enzimática , Inmunosupresores/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Neuropéptido Y/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/agonistas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Wortmanina
14.
Oncogene ; 27(18): 2626-34, 2008 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-17982486

RESUMEN

Transforming growth factor (TGF)-beta1 has a biphasic effect on rat intestinal epithelial (RIE) cells. By itself, TGF-beta1 functions as a tumor suppressor by inhibiting the growth, migration and invasion of RIE cells. We show in this study that in conjunction with epidermal growth factor (EGF), TGF-beta1 helped to augment migration, invasion and anchorage-independent growth (AIG) compared to that by EGF alone. EGF plus TGF-beta1 induced a dramatic morphological change characteristic of epithelial-mesenchymal transition (EMT). The mechanism for this enhanced effect of TGF-beta1 and EGF on oncogenic properties was explored by analysis of EGF- and TGF-beta1-mediated signaling pathways and complementary DNA arrays. TGF-beta1 augmented EGF-mediated signaling of mitogen-activated protein kinase (MAPK) and AKT by enhancing and prolonging the activation of the former and prolonging the activation of the latter. Inhibition of MAPK, but not phosphoinositide-3 kinase (PI3K), abolished TGF-beta1 plus EGF-induced EMT and downregulation of E-cadherin at mRNA and protein levels. By contrast, cell migration and invasion were sensitive to inhibition of either MAPK or PI3 kinase. TGF-beta1 plus EGF-induced AIG was significantly more resistant to inhibition of PI3K and MAPK compared to that induced by EGF alone. EGF and TGF-beta1 synergistically induced the expression of a series of proteases including matrix metalloproteinase (MMP) 1 (collagenase), MMP3, MMP9, MMP10, MMP14 and cathepsin. Among them, the expression of MMP1, MMP3, MMP9 and MMP10 was MAPK dependent. Inhibition of the MMPs or cathepsin significantly blocked EGF plus TGF-beta1-induced invasion, but had no effect on colony formation. Phospholipase C (PLC) and Cox2 induced by EGF plus TGF-beta1 also played a significant role in invasion, whereas PLC was also important for colony formation. Our study reveals specific signaling functions and induction of genes differentially required for enhanced effect of EGF- and TGF-beta1-induced oncogenic properties, and helps to explain the tumor-promoting effect of TGF-beta1 in human cancer with elevated expression or activation of TGF-beta1 and receptor protein tyrosine kinases.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Supresoras de Tumor/agonistas , Animales , Catepsinas/antagonistas & inhibidores , Catepsinas/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Sinergismo Farmacológico , Factor de Crecimiento Epidérmico/agonistas , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/patología , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Factor de Crecimiento Transformador beta1/agonistas , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/farmacología , Fosfolipasas de Tipo C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA