Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 80(1): 136-44, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26991260

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease of premature infants. Connective tissue growth factor (CTGF) plays an important role in tissue development and remodeling. We have previously shown that targeted overexpression of CTGF in alveolar type II epithelial cells results in BPD-like pathology and activates ß-catenin in neonatal mice. METHODS: Utilizing this transgenic mouse model and ICG001, a specific pharmacological inhibitor of ß-catenin, we tested the hypothesis that ß-catenin signaling mediates the effects of CTGF in the neonatal lung. Newborn CTGF mice and control littermates received ICG001 (10 mg/kg/dose) or placebo (dimethyl sulfoxide, equal volume) by daily i.p. injection from postnatal day 5 to 15. Alveolarization, vascular development, and pulmonary hypertension (PH) were analyzed. RESULTS: Administration of ICG001 significantly downregulated expression of cyclin D1, collagen 1a1, and fibronectin, which are the known target genes of ß-catenin signaling in CTGF lungs. Inhibition of ß-catenin signaling improved alveolar and vascular development and decreased pulmonary vascular remodeling. More importantly, the improved vascular development and vascular remodeling led to a decrease in PH. CONCLUSION: ß-Catenin signaling mediates the autocrine and paracrine effects of CTGF in the neonatal lung. Inhibition of CTGF-ß-catenin signaling may provide a novel therapy for BPD.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/genética , Pulmão/metabolismo , Transdução de Sinais , beta Catenina/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Lavagem Broncoalveolar , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibronectinas/metabolismo , Hiperóxia/patologia , Hipertensão Pulmonar/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Alvéolos Pulmonares/patologia , Pirimidinonas/farmacologia , beta Catenina/metabolismo
2.
Invest Ophthalmol Vis Sci ; 53(13): 8076-85, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23139271

RESUMO

PURPOSE: To investigate signal transduction pathways for connective tissue growth factor (CTGF) in human corneal fibroblasts (HCF). METHODS: Expression of 75 kinases in cultures of serum-starved (HCF) were investigated using protein kinase screens, and changes in levels of phosphorylation of 31 different phosphoproteins were determined at 0, 5, and 15 minutes after treatment with CTGF. Levels of phosphorylation of three signal transducing phosphoproteins (extracellular regulated kinase 1 [ERK1], extracellular regulated kinase 2 [ERK2] [MAPKs], and signal transducer and activator of transcription 3 [STAT3]) were measured at nine time points after exposure to CTGF using Western immunoblots. Inhibition of Ras, MEK1/2 (MAPKK), and ERK1/2, on CTGF-stimulated fibroblast proliferation and collagen gel contraction was assessed using selective inhibitors farnesylthiosalicylic acid, PD-98059, and SB203580, respectively. RESULTS: Thirty two of the 75 kinases (43%) evaluated by the kinase screen were detected in extracts of quiescent HCF, suggesting these kinases are available to respond acutely to CTGF exposure. Addition of CTGF increased levels of phosphorylation of five phosphoproteins (ERK1 and 2, MEK1/2 [MAPKK], STAT3, and SAPK/JNK), and decreased levels of phosphorylation of 14 phosphoproteins (including protein kinases B and C) after 5 and 15 minutes. Further analysis of ERK1 and 2 and STAT3 phosphorylation showed rapid increases within 1 minute of CTGF exposure that peaked between 5 and 10 minutes then returned to pretreatment levels by 30 minutes. Treatment of HCF with selective inhibitors of Ras, MEK 1/2, and ERK1/2 individually blocked both CTGF induced cell proliferation, and collagen gel contraction. CONCLUSIONS: Results from protein kinase screens and selective kinase inhibitors demonstrate Ras/MEK/ERK/STAT3 pathway is required for CTGF signaling in HCF.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/farmacologia , Ceratócitos da Córnea/efeitos dos fármacos , Proteínas Quinases/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Ceratócitos da Córnea/enzimologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo
3.
Invest Ophthalmol Vis Sci ; 53(7): 3387-94, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22511630

RESUMO

PURPOSE: To biochemically characterize the receptor for connective tissue growth factor (CTGF) of human corneal fibroblasts (HCF). METHODS: Radiolabeled recombinant human CTGF was used to determine the specificity and time course of binding to low-passage cultures of HCF. The affinity and number of receptors present were calculated by Scatchard and best-fit analyses. In vitro immunoprecipitation assays with radiolabeled CTGF and soluble mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF-2-R) alone, or with CTGF-related growth factors were conducted. Additionally, (125)I-CTGF-binding and CTGF-stimulated proliferation were measured in cultures of M6P/IGF-2-R knockout fibroblasts. RESULTS: Binding of (125)I-CTGF to fibroblast cultures was significantly displaced by CTGF, but not by related growth factors. Scatchard plot analysis indicated the presence of both a high-affinity, low-abundance binding site, and a low-affinity, high-abundance binding site; whereas, the best-fit analysis suggests a single high-affinity, low-abundance binding site. A 280 kDa complex containing cross-linked (125)I-CTGF was immunoprecipitated by antibodies to CTGF or M6P/IGF-2-R. M6P/IGF-2-R knockout cells have a reduced proliferative response to TGF-ß, and don't proliferate at all in response to CTGF. CONCLUSIONS: CTGF binds to the M6P/IGF-2-R with high affinity, and the M6P/IGF-2-R is required for CTGF-stimulated proliferation in fibroblasts. These observations suggest that the M6P/IGF-2-R may be a new antifibrotic target.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Ceratócitos da Córnea/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Autorradiografia , Sítios de Ligação , Western Blotting , Proliferação de Células , Células Cultivadas , Ceratócitos da Córnea/citologia , Fibroblastos , Humanos , Imunoprecipitação , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro
4.
Int Immunopharmacol ; 13(1): 101-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22465472

RESUMO

Sulfur mustard (SM) is a highly toxic chemical warfare agent that remains a threat to human health. The immediate symptoms of pulmonary distress may develop into chronic lung injury characterized by progressive lung fibrosis, the major cause of morbidity among the surviving SM victims. Although SM has been intensely investigated, little is known about the mechanism(s) by which SM induces chronic lung pathology. Increasing evidence suggests that IL-17(+) cells are critical in fibrosis, including lung fibrotic diseases. In this study we exposed F344 rats and cynomolgus monkeys to SM via inhalation and determined the molecular and cellular milieu in their lungs at various times after SM exposure. In rats, SM induced a burst of pro-inflammatory cytokines/chemokines within 72 h, including IL-1ß, TNF-α, IL-2, IL-6, CCL2, CCL3, CCL11, and CXCL1 that was associated with neutrophilic infiltration into the lung. At 2 wks and beyond (chronic phase), lymphocytic infiltration and continued elevated expression of cytokines/chemokines were sustained. TGF-ß, which was undetectable in the acute phase, was strongly upregulated in the chronic phase; these conditions persisted until the animals were sacrificed. The chronic phase was also associated with myofibroblast proliferation, collagen deposition, and presence of IL-17(+) cells. At ≥30 days, SM inhalation promoted the accumulation of IL-17(+) cells in the inflamed areas of monkey lungs. Thus, SM inhalation causes acute and chronic inflammatory responses; the latter is characterized by the presence of TGF-ß, fibrosis, and IL-17(+) cells in the lung. IL-17(+) cells likely play an important role in the pathogenesis of SM-induced lung injury.


Assuntos
Substâncias para a Guerra Química/toxicidade , Exposição por Inalação , Lesão Pulmonar/induzido quimicamente , Gás de Mostarda/toxicidade , Fibrose Pulmonar/induzido quimicamente , Células Th17/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Citocinas/genética , Feminino , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Contagem de Linfócitos , Macaca fascicularis , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Células Th17/imunologia
5.
Proc Natl Acad Sci U S A ; 108(37): 15242-7, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21876171

RESUMO

Type 1 and type 2 diabetes result from an absolute or relative reduction in functional ß-cell mass. One approach to replacing lost ß-cell mass is transplantation of cadaveric islets; however, this approach is limited by lack of adequate donor tissue. Therefore, there is much interest in identifying factors that enhance ß-cell differentiation and proliferation in vivo or in vitro. Connective tissue growth factor (CTGF) is a secreted molecule expressed in endothelial cells, pancreatic ducts, and embryonic ß cells that we previously showed is required for ß-cell proliferation, differentiation, and islet morphogenesis during development. The current study investigated the tissue interactions by which CTGF promotes normal pancreatic islet development. We found that loss of CTGF from either endothelial cells or ß cells results in decreased embryonic ß-cell proliferation, making CTGF unique as an identified ß cell-derived factor that regulates embryonic ß-cell proliferation. Endothelial CTGF inactivation was associated with decreased islet vascularity, highlighting the proposed role of endothelial cells in ß-cell proliferation. Furthermore, CTGF overexpression in ß cells during embryogenesis using an inducible transgenic system increased islet mass at birth by promoting proliferation of immature ß cells, in the absence of changes in islet vascularity. Together, these findings demonstrate that CTGF acts in an autocrine manner during pancreas development and suggest that CTGF has the potential to enhance expansion of immature ß cells in directed differentiation or regeneration protocols.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Animais , Comunicação Autócrina , Linhagem da Célula , Proliferação de Células , Tamanho Celular , Desenvolvimento Embrionário , Camundongos , Modelos Biológicos , Morfogênese , Ratos
6.
J Toxicol Environ Health A ; 74(13): 875-85, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21598172

RESUMO

Sulfur mustard (SM), a vessicating agent, has been used in chemical warfare since 1918. The purpose of this study was to quantitate SM vapor deposition, tissue distribution, and excretion following intratracheal inhalation in rats and cutaneous exposure in guinea pigs. 14C-SM vapors for inhalation studies were generated by metering liquid 14C-SM into a heated J tube. Vapors were transported via carrier air supplemented with oxygen and isoflurane to an exposure plenum. Anesthetized rats with transorally placed tracheal catheters were connected to the plenum port via the catheter hub for exposure (approximately 250 mg 14C-SM vapor/m(3); 10 min). For dermal exposure, 3 Teflon cups (6.6 cm(2) exposure area per cup) were applied to the backs of each animal and vapors (525 mg 14C-SM/m(3); 12 min) were generated by applying 6 µl 14C-SM to filter paper within each cup. Animals were euthanized at selected times up to 7 d postexposure. SM equivalents deposited in rats and guinea pigs were 18.1 ± 3 µg and 29.8 ± 5.31 µg, respectively. Inhaled SM equivalents rapidly distributed throughout the body within 2 h postexposure, with the majority (>70%) of material at that time located in carcass and pelt. In guinea pigs, >90% of deposited SM equivalents remained in skin, with minor distribution to blood and kidneys. Urine was the primary route of excretion for both species. Results indicate inhaled SM is rapidly absorbed from the lung and distributed throughout the body while there is limited systemic distribution following cutaneous exposure.


Assuntos
Substâncias para a Guerra Química/farmacocinética , Gás de Mostarda/farmacocinética , Pele/efeitos dos fármacos , Animais , Gases/farmacocinética , Cobaias , Exposição por Inalação , Intubação Intratraqueal , Rim/química , Pulmão/química , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Gás de Mostarda/análise , Ratos , Ratos Endogâmicos F344 , Pele/química , Pele/metabolismo , Distribuição Tecidual
7.
Toxicol Mech Methods ; 21(7): 547-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21473735

RESUMO

Sulfur mustard (SM) is a chemical threat agent for which the effects have no current treatment. Due to the ease of synthesis and dispersal of this material, the need to develop therapeutics is evident. The present article details the techniques used to develop SM laboratory exposure systems for the development of animal models of ocular and dermal injury. These models are critical to enable evaluation of SM injury and therapeutics against that injury. Iterative trials were conducted to optimize dermal and ocular injury models in guinea pigs and rabbits respectively. The goal was a homogeneous and diffuse ocular and dermal injury that compares to the human injury. Dermal exposures were conducted by either a flow-past or static vapor cup system. Ocular exposures were conducted by a static exposure system. Ocular and dermal exposures were conducted with vaporized SM. Vapor concentrations increased with time in the dermal and ocular exposure systems but were stable with varying amounts of applied SM. A dermal deposition estimation study was also conducted. Deposited volumes increased with exposure time.


Assuntos
Vesícula/induzido quimicamente , Queimaduras Químicas/etiologia , Substâncias para a Guerra Química/toxicidade , Modelos Animais de Doenças , Queimaduras Oculares/etiologia , Gás de Mostarda/toxicidade , Animais , Substâncias para a Guerra Química/farmacocinética , Cobaias , Gás de Mostarda/farmacocinética , Coelhos , Pele/efeitos dos fármacos , Pele/lesões , Pele/metabolismo , Absorção Cutânea , Fatores de Tempo , Volatilização
8.
Wound Repair Regen ; 19(3): 348-57, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21410818

RESUMO

The objective of these studies was to provide detailed analyses of the time course of sulfur mustard (SM) vapor-induced clinical, histological, and biochemical changes following cutaneous exposure in hairless guinea-pigs. Three 6 cm(2) sites on the backs of each guinea-pig were exposed to SM vapor (314 mg(3) ) for 6 minutes (low dose) or 12 minutes (high dose). Animals were killed at 6, 24, and 48 hours, or 2 weeks postexposure. Erythema, edema, histopathology, and analysis of matrix metalloproteinase (MMP)-2 and -9 content were evaluated. Erythema was observed by 6 hours, and edema by 24 hours postexposure. Vapor exposure caused epidermal necrosis with varying degrees of dermatitis, ulceration, hemorrhage, and separation of the dermis from the epidermis. Later changes included epidermal regeneration with hyperplasia and formation of granulation tissue in the dermis with loss of hair follicles and glandular structures. Relative amounts of pro and active MMP-2 and MMP-9 were significantly increased in the high-dose SM group at 2 weeks. Erythema, edema, and histologic changes are consistent with findings among human victims of SM attack. This model, with observations to 2 weeks, will be useful in assessing the efficacy of countermeasures against SM.


Assuntos
Dermatite de Contato/patologia , Fármacos Dermatológicos/toxicidade , Eritema/induzido quimicamente , Gás de Mostarda/toxicidade , Animais , Queimaduras Químicas/patologia , Modelos Animais de Doenças , Edema/induzido quimicamente , Cobaias , Masculino , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Necrose , Pele/efeitos dos fármacos , Pele/enzimologia , Pele/patologia , Fatores de Tempo , Volatilização
9.
Inhal Toxicol ; 22(11): 896-902, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20569120

RESUMO

CONTEXT: Sulfur mustard (SM) causes skin blistering and long-term pulmonary dysfunction. Its adverse effects have been studied in battlefield-exposed humans, but lack of knowledge regarding confounding factors makes interpretation challenging. Animal studies are critical to understanding mechanisms, but differences between animals and humans must be addressed. Studies of cultured human cells can bridge animal studies and humans. OBJECTIVE: Evaluate effects of SM vapor on airway cells. MATERIALS AND METHODS: We examined responses of differentiated human tracheal/bronchial epithelial cells, cultured at an air-liquid interface, to SM vapors. SM effects on metabolic activity (Water Soluble Tetrazolium (WST) assay), cytokine and metalloproteinase secretion, and cellular heme oxygenase 1 (HO-1), an oxidative stress indicator, were measured after 24 h. RESULTS: At noncytotoxic levels of exposure, interleukin 8 and matrix metalloproteinase-13 were significantly increased in these cultures, but HO-1 was not significantly affected. DISCUSSION AND CONCLUSION: Exposure of differentiated airway epithelial cells to sub-cytotoxic levels of SM vapor induced inflammatory and degradative responses that could contribute to the adverse health effects of inhaled SM.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Gás de Mostarda/administração & dosagem , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Cultivadas , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Heme Oxigenase-1/metabolismo , Humanos , Gás de Mostarda/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Mucosa Respiratória/citologia , Traqueia/citologia , Traqueia/efeitos dos fármacos , Traqueia/metabolismo , Volatilização
10.
Int Immunopharmacol ; 10(2): 193-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19887117

RESUMO

Sulfur mustard (SM, bis-(2-chloroethyl) sulfide) is a well known chemical warfare agent that may cause long-term debilitating injury. Because of the ease of production and storage, it has a strong potential for chemical terrorism; however, the mechanism by which SM causes chronic tissue damage is essentially unknown. SM is a potent protein alkylating agent, and we tested the possibility that SM modifies cellular antigens, leading to an immunological response to "altered self" and a potential long-term injury. To that end, in this communication, we show that dermal exposure of euthymic hairless guinea pigs induced infiltration of both CD4(+) and CD8(+) T cells into the SM-exposed skin and strong upregulated expression of proinflammatory cytokines and chemokines (TNF-alpha, IFN-gamma, and IL-8) in distal tissues such as the lung and the lymph nodes. Moreover, we present evidence for the first time that SM induces a specific delayed-type hypersensitivity response that is associated with splenomegaly, lymphadenopathy, and proliferation of cells in these tissues. These results clearly suggest that dermal exposure to SM leads to immune activation, infiltration of T cells into the SM-exposed skin, delayed-type hypersensitivity response, and molecular imprints of inflammation in tissues distal from the site of SM exposure. These immunological responses may contribute to the long-term sequelae of SM toxicity.


Assuntos
Substâncias para a Guerra Química/toxicidade , Hipersensibilidade Tardia/induzido quimicamente , Gás de Mostarda/toxicidade , Pele/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Cobaias , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/patologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Pulmão/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Masculino , Pele/imunologia
11.
Toxicol Mech Methods ; 20(1): 14-24, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20025432

RESUMO

Sulfur mustard (SM) is a chemical threat agent for which its effects have no current treatment. Due to the ease of synthesis and dispersal of this material, the need to develop therapeutics is evident. The present manuscript details the techniques used to develop SM laboratory exposure systems for the development of animal models of pulmonary injury. These models are critical for evaluating SM injury and developing therapeutics against that injury. Iterative trials were conducted to optimize a lung injury model. The resulting pathology was used as a guide, with a goal of effecting homogeneous and diffuse lung injury comparable to that of human injury. Inhalation exposures were conducted by either nose-only inhalation or intubated inhalation. The exposures were conducted to either directly vaporized SM or SM that was nebulized from an ethanol solution. Inhalation of SM by nose-only inhalation resulted in severe nasal epithelial degeneration and minimal lung injury. The reactivity of SM did not permit it to transit past the upper airways to promote lower airway injury. Intratracheal inhalation of SM vapors at a concentration of 5400 mg x min/m(3) resulted in homogeneous lung injury with no nasal degeneration.


Assuntos
Substâncias para a Guerra Química/toxicidade , Modelos Animais de Doenças , Pneumopatias/induzido quimicamente , Pulmão/efeitos dos fármacos , Gás de Mostarda/toxicidade , Aerossóis , Animais , Feminino , Exposição por Inalação , Intubação Intratraqueal , Pulmão/patologia , Pneumopatias/patologia , Tamanho da Partícula , Projetos Piloto , Ratos , Ratos Endogâmicos F344 , Conchas Nasais/efeitos dos fármacos , Conchas Nasais/patologia , Volatilização
12.
Am J Respir Cell Mol Biol ; 36(5): 552-61, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17197570

RESUMO

Transforming growth factor-beta1 (TGF-beta1) has been implicated as a major negative regulator of lung branching morphogenesis. Since connective tissue growth factor (CTGF) is a downstream mediator of TGF-beta1 effects on mesenchymal cells, we hypothesized that TGF-beta1 induces CTGF expression in mouse embryonic lung explants and that CTGF mediates TGF-beta1 inhibition of branching morphogenesis. We show that addition of TGF-beta1 to the serum-free medium of embryonic day (E)12.5 lung explant cultures inhibited branching morphogenesis and induced CTGF mRNA expression in time- and dose-dependent manners. In contrast to basal endogenous CTGF protein, which was exclusively localized in the distal airway epithelium, TGF-beta1-induced CTGF protein was localized in both the epithelium and the mesenchyme. Addition of exogenous CTGF to culture medium directly inhibited branching morphogenesis. To identify the signal transduction pathway through which TGF-beta1 induces CTGF, we used SB431542, a specific inhibitor for TGF-beta type I receptor (TbetaRI)/ALK-5 to block TGF-beta1-induced Smad2/3 phosphorylation. Consequently, SB431542 stimulated normal branching morphogenesis and blocked TGF-beta1 inhibition of branching. Furthermore, SB-431542 blocked both endogenous and TGF-beta1-induced expression of CTGF mRNA and protein. These results demonstrate for the first time that TGF-beta1 induces CTGF expression in mouse embryonic lung explants, that CTGF inhibits branching morphogenesis, and that both endogenous and TGF-beta1-induced CTGF expression are mediated by the TbetaRI/ALK-5-dependent Smad2 signaling pathway.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Receptores de Ativinas Tipo I/antagonistas & inibidores , Animais , Benzamidas/farmacologia , Fator de Crescimento do Tecido Conjuntivo , Cicloeximida/farmacologia , Dioxóis/farmacologia , Feminino , Proteínas Imediatamente Precoces/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Pulmão/citologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Morfogênese/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Proteína Smad2/metabolismo
13.
FASEB J ; 19(7): 729-38, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15857887

RESUMO

All members of the Ctgf, Cyr61, and Nov (CCN) family share a high degree of sequence homology and conservation of structural motifs and domains. Here, we present data about a structure function analysis of connective tissue growth factor (CTGF), a prototypic member of the CCN family, which has been shown to be a downstream mediator of transforming growth factor-beta activities on fibroblasts. Our findings demonstrate the two domains of CTGF function to mediate two distinct biological effects. The N-terminal domain of CTGF mediates myofibroblast differentiation and collagen synthesis. The C-terminal domain of CTGF mediates fibroblast proliferation. These data provide a molecular basis for the divergence of CTGF actions on connective tissue cell types and suggest a model for functional analysis of all of the CCN family gene products.


Assuntos
Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Fibroblastos/citologia , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Células Musculares/citologia , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Colágeno/biossíntese , Células do Tecido Conjuntivo , Fator de Crescimento do Tecido Conjuntivo , Glicoconjugados/metabolismo , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/química , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Rim , Proteína Sobre-Expressa em Nefroblastoma , Ratos , Proteínas Recombinantes , Transdução de Sinais , Somatomedinas/metabolismo , Relação Estrutura-Atividade , Sulfatos/metabolismo , Trombospondina 1/química , Fator de Crescimento Transformador beta/fisiologia
14.
FASEB J ; 19(2): 270-1, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15579670

RESUMO

The vasculature consists of endothelial cells (ECs) lined by pericyte/vascular smooth muscle cells (vSMCs). Pericyte/vSMCs provide support to the mature vasculature but are also essential for normal blood vessel development. To determine how pericyte-EC communication influences vascular development, we used the well-established in vitro model of TGFbeta-stimulated differentiation of 10T1/2 cells into pericyte/vSMCs. Microarray analysis was performed to identify genes that were differentially expressed by induced vs. uninduced 10T1/2 cells. We discovered that these cells show an angiogenic program of gene expression, with up-regulation of several genes previously implicated in angiogenesis, including VEGF, IL-6, VEGF-C, HB-EGF, CTGF, tenascin C, integrin alpha5, and Eph receptor A2. Up-regulation of some genes was validated by Western blots and immunocytochemistry. We also examined the functional significance of these gene expression changes. VEGF and IL-6 alone and in combination were important in 10T1/2 cell differentiation. Furthermore, we used a coculture system of 10T1/2 and human umbilical vein ECs (HUVECs), resulting in the formation of cordlike structures by the HUVECs. This cordlike structure formation was disrupted when neutralizing antibodies to VEGF or IL-6 were added to the coculture system. The results of these studies show that factors produced by pericytes may be responsible for recruiting ECs and promoting angiogenesis. Therefore, a further understanding of the genes involved in pericyte differentiation could provide a novel approach for developing anti-angiogenic therapies.


Assuntos
Diferenciação Celular/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Análise em Microsséries/métodos , Neovascularização Fisiológica/genética , Pericitos/citologia , Animais , Linhagem Celular , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Genes/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C3H , Células-Tronco Multipotentes/química , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Pericitos/química , Pericitos/metabolismo , Veias Umbilicais/química , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
15.
Invest Ophthalmol Vis Sci ; 45(4): 1109-16, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15037576

RESUMO

PURPOSE: This study was undertaken to investigate the role of connective tissue growth factor (CTGF) in fibroblast-to-myofibroblast differentiation and fibroblast-mediated collagen matrix contraction in the presence of mechanical stress. METHODS: An in vitro three-dimensional contraction model of human corneal-fibroblast-seeded collagen lattices (FSCLs) in the presence of mechanical stress generated by attaching the lattices to the culture well was used to measure FSCL contraction. FSCLs were treated with CTGF; TGF-beta1; serum-free (SF) control medium; or TGF-beta1 plus antisense oligodeoxynucleotides to CTGF; TGF-beta1 plus scrambled-sequence oligodeoxynucleotide to CTGF; or TGF-beta antibody. Expression of alpha-smooth muscle actin (alpha-SMA) by fibroblasts in FSCLs was detected by immunostaining and confocal microscopy, whereas ELISA was used for the fibroblasts cultured on plastic. The conditioned media were analyzed by ELISA for CTGF production. RESULTS: Exogenous CTGF stimulated significantly less collagen matrix contraction and myofibroblast differentiation than TGF-beta1, but similar to that stimulated by SF. TGF-beta1 stimulated fibroblasts to express CTGF. CTGF antisense oligodeoxynucleotide inhibited TGF-beta1-stimulated myofibroblast differentiation and FSCL contraction. Exogenous CTGF circumvented the inhibitory effects of CTGF antisense on FSCL contraction. TGF-beta antibody significantly inhibited FSCL contraction and myofibroblast differentiation under mechanical stress and SF control conditions. CONCLUSIONS: In the presence of mechanical stress, CTGF is necessary for TGF-beta1-stimulation of myofibroblast differentiation and subsequent collagen matrix contraction, but CTGF alone is not sufficient to induce myofibroblast differentiation and collagen matrix contraction. Thus, TGF-beta1 appears to regulate multiple genes that are essential for fibroblast-mediated contraction of stressed matrix, one of which is CTGF.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colágeno/metabolismo , Córnea/citologia , Matriz Extracelular/metabolismo , Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Estresse Mecânico , Fator de Crescimento Transformador beta/farmacologia , Actinas/metabolismo , Técnicas de Cultura de Células , Fator de Crescimento do Tecido Conjuntivo , Córnea/metabolismo , Ensaio de Imunoadsorção Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/farmacologia , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Microscopia Confocal , Oligonucleotídeos Antissenso/farmacologia , Fator de Crescimento Transformador beta1
16.
FASEB J ; 18(3): 469-79, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15003992

RESUMO

Fibroblast proliferation, differentiation into myofibroblasts, and increased collagen synthesis are key events during both normal wound repair and fibrotic lesion formation. Here we report that these biological responses to TGF-beta by fibroblasts are regulated via a CTGF-dependent pathway in concert with either EGF or IGF-2. Our studies indicate these responses to TGF-beta are mutually exclusive, and cells that are proliferating do not express alpha-SMA or elevated levels of collagen synthesis. Cells expressing alpha-SMA do not exhibit DNA synthesis but do coexpress higher levels of types I and III collagen mRNA. Thus, fibroblast proliferation and differentiation are controlled by combinatorial signaling pathways involving not only components of the TGF-beta/CTGF pathway, but also signaling events induced by EGF and IGF-2-activated receptors. Collectively, our studies indicate TGF-beta functions as a classic embryonic inducer, initiating a cascade that is controlled by other factors in the cellular environment. We propose a model for this process with regard to wound repair and fibrotic lesion formation that is likely applicable to other instances of CTGF action during embryogenesis.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Fibroblastos/citologia , Proteínas Imediatamente Precoces/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mioblastos/citologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Cicatrização/fisiologia , Actinas/biossíntese , Actinas/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular/citologia , Linhagem Celular/efeitos dos fármacos , Colágeno/biossíntese , Colágeno/genética , Fator de Crescimento do Tecido Conjuntivo , Replicação do DNA/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Rim , RNA Mensageiro/biossíntese , Ratos , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
17.
Invest Ophthalmol Vis Sci ; 45(2): 485-91, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14744889

RESUMO

PURPOSE: Connective tissue growth factor (CTGF) appears to play a significant role in mediating fibrosis in several tissues. To gain further understanding of the role of CTGF in the scar formation that occurs after glaucoma filtering surgery (GFS), experiments were performed in a rabbit model. METHODS: . Three experiments were performed: (1) CTGF and transforming growth factor (TGF)-beta expression were measured quantitatively after GFS, using ELISA. (2) After GFS conjunctival bleb tissues were immunostained for the presence of CTGF and TGF-beta. (3) Exogenous CTGF was injected into mitomycin-C (MMC)-treated filtering blebs and the scaring response compared to TGF-beta and physiological saline-injected blebs. RESULTS: CTGF and TGF-beta were expressed maximally by day 5 after surgery and were both shown to be present in the bleb tissues after GFS. The addition of exogenous CTGF and TGF-beta increased the rate of failure of GFS blebs. CONCLUSIONS: These data support the hypothesis that CTGF plays an important role in scarring and wound contracture after GFS. Inhibition of CTGF synthesis or its action may help prevent bleb failure and improve long-term GFS outcomes.


Assuntos
Túnica Conjuntiva/metabolismo , Cirurgia Filtrante , Glaucoma/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Malha Trabecular/metabolismo , Animais , Túnica Conjuntiva/efeitos dos fármacos , Túnica Conjuntiva/patologia , Fator de Crescimento do Tecido Conjuntivo , Ensaio de Imunoadsorção Enzimática , Glaucoma/cirurgia , Proteínas Imediatamente Precoces/farmacologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Modelos Animais , Coelhos , Malha Trabecular/efeitos dos fármacos , Malha Trabecular/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia
18.
Am J Pathol ; 163(5): 2043-52, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14578203

RESUMO

Excessive cell-mediated tissue contraction after injury can lead to morbid contractile scarring in the body. In the eye this can cause blindness because of posterior capsule opacification, proliferative vitroretinopathy, failure of glaucoma filtration surgery, and corneal haze. During repair, transforming growth factor (TGF)-beta and connective tissue growth factor (CTGF) genes are co-ordinately expressed. Although TGF-beta and CTGF stimulate new matrix deposition, their role and regulation during contractile scarring is unknown. In this study, an in vitro model of collagen matrix contraction culminating from tractional forces generated by fibroblasts showed that both TGF-beta(1) and CTGF-stimulated contraction. Using a specific anti-sense oligodeoxynucleotide to CTGF the procontractile activity of TGF-beta(1) was found to be mediated by CTGF. During contraction fibroblasts produced similar levels of matrix metalloproteinases (MMPs)-2 and -9 with TGF-beta(1) or CTGF and a modest increase in MMP-1 with CTGF only (indicated by zymography and enzyme-linked immunosorbent assay). The requirement of MMPs for contraction was demonstrated using a broad-spectrum synthetic inhibitor. This study demonstrates a new function for CTGF in mediating matrix contraction by fibroblasts involving MMPs and suggests a novel regulatory mechanism for TGF-beta-stimulated contraction. Inhibition of CTGF activity or gene transcription could be a suitable target for anti-scarring therapies.


Assuntos
Cicatriz/metabolismo , Matriz Extracelular/efeitos dos fármacos , Fibroblastos/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Northern Blotting , Células Cultivadas , Colágeno/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Fibroblastos/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/efeitos dos fármacos , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/análise , Óxidos de Enxofre , Transcrição Gênica , Fator de Crescimento Transformador beta1
19.
Invest Ophthalmol Vis Sci ; 44(5): 1879-87, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12714619

RESUMO

PURPOSE: Connective tissue growth factor (CTGF) has been linked to fibrosis in several tissues. In this study, the interactions between CTGF and transforming growth factor (TGF)-beta were assessed in human corneal fibroblasts, and the levels and location of CTGF protein and mRNA were measured during healing of excimer laser ablation wounds in rat corneas. METHODS: Human corneal fibroblasts were incubated with TGF-beta1, -beta2, and -beta3 isoforms, and CTGF mRNA and protein were measured. CTGF was immunolocalized in the cultured fibroblasts by using a specific antibody. Regulation of collagen synthesis by TGF-beta and CTGF was assessed in human corneal fibroblasts with a neutralizing antibody and an antisense oligonucleotide to CTGF. CTGF mRNA and protein were measured in rat corneas up to day 21 after excimer ablation of the cornea. CTGF protein was immunolocalized in rat corneas after photorefractive keratectomy (PRK), and the presence of CTGF mRNA and protein in ex vivo rat corneal scrapings was established. RESULTS: All three TGF-beta isoforms stimulated expression of CTGF in human corneal fibroblasts, and CTGF was immunolocalized in the cells. Both TGF-beta and CTGF increased collagen synthesis in corneal fibroblasts. Furthermore, CTGF antibody or antisense oligonucleotide blocked TGF-beta-stimulated collagen synthesis. CTGF protein and mRNA increased in rat corneas through day 21 after PRK. CTGF expression was also detected in ex vivo scrapings of rat corneas. CONCLUSIONS: These data demonstrate that CTGF is expressed by corneal cells after stimulation by TGF-beta, that CTGF expression increases significantly during corneal wound healing, and that CTGF mediates the effects of TGF-beta induction of collagen synthesis by corneal fibroblasts. These data support the hypothesis that CTGF promotes corneal scar formation and imply that regulating CTGF synthesis and action may be an important goal for reducing corneal scarring.


Assuntos
Córnea/metabolismo , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ceratectomia Fotorrefrativa , Cicatrização/fisiologia , Animais , Técnicas de Cultura de Células , Colágeno/biossíntese , Fator de Crescimento do Tecido Conjuntivo , Córnea/citologia , Córnea/efeitos dos fármacos , Córnea/cirurgia , Ensaio de Imunoadsorção Enzimática , Fibroblastos/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/farmacologia , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Lasers de Excimer , Oligonucleotídeos Antissenso/farmacologia , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia
20.
Arthritis Rheum ; 48(3): 798-806, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12632435

RESUMO

OBJECTIVE: To examine the mechanism of collagen induction by connective tissue growth factor (CTGF), a profibrotic cytokine overexpressed in the skin of patients with systemic sclerosis (SSc). METHODS: Dermal fibroblasts from 7 SSc patients and 7 matched healthy adult donors were stimulated with CTGF in the presence or absence of the culture-medium supplement, insulin-transferrin-selenium (ITS). Expression of collagen protein was analyzed by a (3)H-proline incorporation assay. To identify the signaling pathways mediating CTGF induction of collagen, pharmacologic inhibitors were used, including rottlerin, a protein kinase C delta (PKC delta) inhibitor. RESULTS: Collagen levels in both SSc and normal fibroblasts were increased after treatment with transforming growth factor beta in serum-free medium, whereas no stimulation was observed following addition of CTGF. In the presence of ITS, CTGF (2.5 ng/ml) potently stimulated collagenous protein levels in SSc cell lines (n = 5); however, CTGF was not stimulatory in the majority of normal fibroblasts (n = 6). ITS alone induced collagen levels in normal fibroblasts to the levels observed in SSc skin fibroblasts, thereby diminishing the hallmark difference in basal collagen levels in these cell types. Insulin was the ITS component responsible for promoting the basal and CTGF stimulation of collagenous proteins. Rottlerin, the PKC delta inhibitor, down-regulated collagen synthesis in normal and SSc fibroblasts cultured in ITS, and inhibited the stimulatory effects of CTGF in cooperation with insulin or of insulin (500 ng/ml) alone. CONCLUSION: Increased responsiveness of SSc fibroblasts to CTGF-mediated collagen synthesis requires the costimulatory activation of insulin signaling pathways to induce matrix production. Blockade of this effect via rottlerin may suggest that PKC delta is a downstream signaling molecule necessary for CTGF stimulation of collagen synthesis.


Assuntos
Colágeno/biossíntese , Fibroblastos/metabolismo , Proteínas Imediatamente Precoces/farmacologia , Insulina/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Escleroderma Sistêmico/metabolismo , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Recém-Nascido , Prolina/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Escleroderma Sistêmico/patologia , Selênio/farmacologia , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Transferrina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...