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










Base de dados
Intervalo de ano de publicação
1.
Curr Med Chem ; 16(11): 1400-17, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19355895

RESUMO

Idiopathic Pulmonary Fibrosis (IPF) is characterized by injury and loss of lung epithelial cells, accumulation of fibroblasts/myofibroblasts and abnormal remodeling of the lung parenchyma. The prognosis for IPF patients is poor and current therapies are largely ineffective in preventing respiratory failure. Current therapeutic approaches target epithelial cell replacement, manipulation of fibroblasts/myofibroblasts, modulation of procoagulant/fibrinolytic activities, cytokine and growth factor production, angiogenesis, and reduction of oxidative stress. Myofibroblasts are the primary effector cells in fibrosis. These cells may be derived by the activation and proliferation of resident lung fibroblasts, from epithelial-mesenchymal transition (EMT), or through recruitment of circulating fibrocytes. Transforming growth factor beta (TGFbeta) is a profibrotic factor that increases fibroblast proliferation, stimulates the synthesis and deposition of connective tissue, and inhibits connective tissue breakdown. TGFbeta acts through the promoter of the type 1 collagen gene causing increased collagen synthesis. In addition, TGFbeta induces EMT in alveolar epithelial cells (AECs) in vitro and in vivo. AECs exhibit substantial plasticity and may serve as a source of fibroblasts and/or myofibroblasts in lung fibrosis. Therapeutic interventions interfering with the pathways that lead to myofibroblast expansion and AEC apoptosis should be of considerable benefit in the treatment of IPF. This review will focus on the critical role of TGFbeta on AECs EMT and myofibroblasts in the development of fibrosis.


Assuntos
Células Epiteliais/patologia , Fibroblastos/patologia , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/fisiopatologia , Alvéolos Pulmonares/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética
2.
J Pathol ; 212(2): 180-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17492827

RESUMO

Myofibroblasts play an essential role in the abnormal deposition of extracellular matrix in pulmonary fibrosis. The presence or prolonged survival of these cells may be a key factor in the pathogenesis of progressive pulmonary fibrosis. Found in inflammatory zone (FIZZ)1 can induce myofibroblast differentiation and has an antiapoptotic effect on embryonic lung explant cultures. In this study, we investigated whether FIZZ1 also has an antiapoptotic effect on mouse lung fibroblasts (MLFs). Cells were treated with FIZZ1 for 24 h and then apoptosis was induced by TNFalpha in the presence of cycloheximide (CHX). FIZZ1 exhibited an antiapoptotic effect in MLFs, as assessed by flow cytometric analysis and TUNEL staining. Moreover, the cell number was higher in the FIZZ1-treated group relative to the non-treated control group after treatment with TNFalpha and CHX. FIZZ1 treatment also inhibited the apoptotic agent-induced activities of caspase-3 and caspase-8. Examination of potential signalling pathways revealed that FIZZ1 induced rapid phosphorylation of ERK-1/2, while PD98059, a MEK/ERK inhibitor, markedly induced activation of caspase-3. This anti-apoptotic effect of FIZZ1 was associated with induction of myofibroblast differentiation in response to FIZZ1 stimulation. Taken together, these findings suggest that FIZZ1 is involved in pulmonary fibrosis through both induction of myofibroblast differentiation and increased or prolonged survival of myofibroblasts. This effect of FIZZ1 was mediated by inhibition of caspase-3 and -8, with involvement of the ERK pathway.


Assuntos
Apoptose/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Proteínas/farmacologia , Animais , Apoptose/imunologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Diferenciação Celular/efeitos dos fármacos , Cicloeximida/farmacologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Fibroblastos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos CBA , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Crescimento Neural/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-akt/análise , Fator de Necrose Tumoral alfa/farmacologia
3.
J Pathol ; 211(4): 455-62, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17177178

RESUMO

Pulmonary fibrosis is characterized by inflammation, genesis of myofibroblasts, and abnormal tissue repair. Despite extensive research, its pathogenesis remains incompletely understood. Previously, the transcription factor CCAAT/enhancer binding protein beta (C/EBPbeta) was found to be a key regulator of myofibroblast differentiation in vitro, and to be involved in the acute phase and inflammatory responses. In an attempt to test the role of C/EBPbeta in the development of pulmonary fibrosis, experiments using C/EBPbeta null mice and their wild-type littermates were conducted. Our findings indicated that, compared to wild-type mice, animals deficient in C/EBPbeta showed significantly reduced fibrotic lesions and collagen deposition in the lung upon endotracheal injection of bleomycin. Further studies on the mechanisms by which C/EBPbeta regulates fibrosis indicated that knockout of C/EBPbeta attenuates inflammatory cytokine expression in bleomycin-treated mice. The reduced alpha-smooth muscle actin gene expression in either isolated lung fibroblasts or lung tissue from bleomycin or saline-treated C/EBPbeta deficient mice suggests that C/EBPbeta regulates myofibroblast differentiation during fibrosis. Consistent with this finding, cells from C/EBPbeta deficient mice exhibited higher proliferative rates than those from wild-type mice. These data suggest that C/EBPbeta plays an essential role in pulmonary fibrosis and that this role appears to be multifactorial with respect to cytokine expression, cell differentiation, and proliferation.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/análise , Fibrose Pulmonar/metabolismo , Actinas , Animais , Antibióticos Antineoplásicos , Bleomicina , Proteína beta Intensificadora de Ligação a CCAAT/deficiência , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Colágeno/análise , Citocinas/análise , Fibroblastos/fisiologia , Expressão Gênica , Genótipo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/metabolismo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , RNA Mensageiro/análise
4.
Oncogene ; 25(7): 1030-41, 2006 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-16205635

RESUMO

Telomerase is induced in certain pathological conditions such as cancer and tissue injury and repair. This induction in fibroblasts from injured lung is repressed by transforming growth factor beta (TGFbeta) via yet unknown mechanisms. In this study, the role of Smad3 in the inhibition of telomerase reverse transcriptase (TERT) gene transcription by TGFbeta was investigated. The rat TERT (rTERT) gene promoter was cloned by PCR amplification and fused with a luciferase reporter gene. This construct was used to analyse regulation of promoter activity in fibroblasts isolated from bleomycin-injured lung with induced telomerase activity. The results showed that TGFbeta inhibited rTERT transcription while stimulating Smad3 expression. Interestingly, TGFbeta also inhibited the expression of c-myc. Cotransfection with a Smad3 expressing plasmid further repressed rTERT transcription and c-myc expression, while cotransfection with the corresponding antisense Smad3 construct had the opposite effect. Mutation of an E-box in the rTERT promoter suppressed its activity, which could be further reduced by TGFbeta treatment. In contrast, mutation at a Smad binding element enhanced promoter activity whose inhibition was impaired by TGFbeta treatment. Thus TGFbeta inhibition of rTERT gene expression was directly mediated by Smad3 via the Smad binding element, while c-myc appears to primarily regulate its constitutive or induced expression.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Pulmão/enzimologia , Pulmão/patologia , Proteína Smad3/metabolismo , Telomerase/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia , Animais , Sequência de Bases , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/enzimologia , Fibrose , Expressão Gênica , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos , Ratos Endogâmicos F344 , Elementos de Resposta , Telomerase/genética
5.
Curr Pharm Des ; 11(30): 3943-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16305523

RESUMO

Pulmonary fibrosis is characterized by lung inflammation and abnormal tissue repair, resulting in the replacement of normal functional tissue with an abnormal accumulation of fibroblasts and deposition of collagen in the lung. This process involves cellular interactions via a complex cytokine-signaling mechanism and heightened collagen gene expression, ultimately resulting in its abnormal collagen deposition in the lung. Our current understanding of the pathogenesis of pulmonary fibrosis suggests that in addition to inflammatory cells, the fibroblast and signaling events that mediate fibroblast proliferation and myofibroblasts, play important roles in the diverse processes that constitute fibrosis. Increasing knowledge of cytokine biology, cytokine-signaling and cell matrix interactions have shed some light on the genesis of pulmonary fibrosis; however, the importance of inflammation in pulmonary fibrosis remains controversial. This remains true because the inflammatory component is variable at the time of diagnosis, and the most potent anti-inflammatory drugs that have been widely used in the treatment of pulmonary fibrosis do not seem to interfere with the fibrotic disease progression. Pulmonary fibrosis is a highly lethal disorder, which continues to pose major clinical challenges because an effective therapeutic regimen is yet to be determined. This review summarizes recent progress in understanding the molecular mechanisms of pulmonary fibrosis, and includes a more detailed discussion of the potential points of therapeutic attack in pulmonary fibrosis. In addition, a detailed discussion is presented regarding each of the potential therapies which have emerged from the animal models of pulmonary fibrosis, and which have been developed through advances in cellular and molecular biology.


Assuntos
Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/fisiopatologia , Animais , Apoptose/fisiologia , Quimiocinas/fisiologia , Citocinas/fisiologia , Substâncias de Crescimento/fisiologia , Humanos , Transplante de Pulmão , Inibidores de Metaloproteinases de Matriz , Neovascularização Fisiológica/efeitos dos fármacos , Fibrose Pulmonar/patologia , Fibrose Pulmonar/cirurgia
7.
Cytokine ; 15(3): 138-47, 2001 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-11554783

RESUMO

Interleukin-4 (IL-4) is known to activate mononuclear cells as well as fibroblasts, all of which are important in the pathogenesis of pulmonary fibrosis. To investigate the potential role of this cytokine, lung IL-4 expression was examined in a murine model of bleomycin-induced pulmonary fibrosis. Lung fibrosis was induced in CBA/J mice by endotracheal injection of bleomycin on day 0. On selected days after treatment, lungs were harvested for reverse transcriptase polymerase chain reaction (RT-PCR), Northern, in-situ hybridization and immunohistochemical analyses. RT-PCR and Northern analyses revealed significant increases in lung IL-4 mRNA content between days 3 and 14 after induction of lung injury, which decreased toward control level after day 21. Both in-situ hybridization and immunohistochemistry showed low or undetectable IL-4 expression in control lungs and in injured lungs before day 3 after bleomycin injection. There was however elevated expression in mononuclear cells and macrophages between days 3 and 14, localized to areas of active fibrosis. These results demonstrate that IL-4 is upregulated significantly in this model. They suggest a potential role for this cytokine in pulmonary fibrosis, perhaps via its ability to stimulate and amplify the inflammatory response, stimulate collagen synthesis in fibroblasts, and thus promote the progression to fibrosis and end stage lung disease.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Fibrose/induzido quimicamente , Interleucina-4/biossíntese , Pulmão/metabolismo , Animais , Northern Blotting , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Cinética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos CBA , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Regulação para Cima
8.
Curr Pharm Des ; 7(11): 1083-103, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472255

RESUMO

Cytokines are critical to a myriad of fundamental homeostatic and pathophysiological processes such as fever, wound healing, inflammation, tissue repair and fibrosis. They play important roles in regulating cell function such as proliferation, migration, and matrix synthesis. It is the balance or the net effect of the complex interplay between these mediators, which appears to play a major role in regulating the initiation, progression and resolution of wounds. Wound healing involves a complex process including induction of acute inflammation by the initial injury, followed by parenchymal and mesenchymal cell proliferation, migration, and activation with production and deposition of extracellular matrix. Failure to resolve or abnormal wound healing results in fibrosis. The latter process involves similar cellular interactions via complex cytokine networks, which result in extensive remodeling with heightened extracellular matrix production and their abnormal deposition in the tissue. Various cytokines, both promoting and inhibiting fibrogenesis, have been implicated in the pathogenesis of fibrosis and wound healing. Recent progress in understanding the mechanisms underlying the pathogenesis of fibrosis leads us to expect that inhibitors of pro-fibrogenic cytokines and growth factors may be useful as novel therapeutic agents in controlling undesirable fibrosis. In this review, the role of cytokines in wound healing and fibrosis will be summarized and highlighted with more detailed discussion reserved for the possible points of therapeutic attack in pulmonary fibrosis. In this review, the major cytokines that are in current clinical use will be also discussed. In addition, advances in the application of novel cytokines and anti-cytokines for accelerating wound healing and attenuating fibrosis both at the experimental and the clinical trial levels will be discussed.


Assuntos
Citocinas/fisiologia , Citocinas/uso terapêutico , Fibrose/terapia , Cicatrização/fisiologia , Animais , Comunicação Celular/fisiologia , Divisão Celular/fisiologia , Matriz Extracelular/fisiologia , Fibrose/etiologia , Fibrose/fisiopatologia , Humanos , Fator de Necrose Tumoral alfa/fisiologia , Cicatrização/efeitos dos fármacos
9.
Int J Biochem Cell Biol ; 33(7): 723-34, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11390280

RESUMO

Myofibroblasts express alpha-smooth muscle actin and have a phenotype intermediate between fibroblasts and smooth muscle cells. Their emergence can be induced by cytokines such as transforming growth factor beta; but the regulatory mechanism for induction of alpha-smooth muscle actin gene expression in myofibroblast differentiation has not been determined. To examine this mechanism at the level of the alpha-smooth muscle actin promoter, rat lung fibroblasts were transfected with varying lengths of the alpha-smooth muscle actin promoter linked to the chloramphenicol acetyl transferase reporter gene and treated with transforming growth factor beta1. The results show that the shortest inducible promoter was 150 base pairs long, suggesting the presence in this region of cis-elements of potential importance in transforming growth factor beta1 induced myofibroblast differentiation. Transfection of "decoy" oligonucleotides corresponding to sequences for four suspected regulatory factors demonstrated that only the transforming growth factor beta control element is involved in the regulation of transforming growth factor beta1-induced alpha-smooth muscle actin expression in myofibroblast differentiation. Consistent with this conclusion is the finding that a mutation in the transforming growth factor beta control element caused a significant reduction in promoter activity. These observations taken together show that alpha-smooth muscle actin promoter regulation during myofibroblast differentiation is uniquely different from that in smooth muscle cells and other cell lines. Since myofibroblasts play a key role in wound contraction and synthesis of extracellular matrix, clarification of this differentiation mechanism should provide new insight into fibrogenesis and suggest future novel strategies for modulation of wound healing and controlling fibrosis.


Assuntos
Actinas/genética , Fibroblastos/citologia , Regulação da Expressão Gênica , Músculo Liso , Proteínas Nucleares , Regiões Promotoras Genéticas , Fator de Crescimento Transformador beta/farmacologia , Actinas/biossíntese , Animais , Diferenciação Celular , Células Cultivadas , Cloranfenicol O-Acetiltransferase/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Pulmão/citologia , Músculo Liso/citologia , Ratos , Elementos de Resposta , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/metabolismo , Transfecção , Fator de Crescimento Transformador beta1
10.
Am J Respir Cell Mol Biol ; 23(4): 460-5, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11017910

RESUMO

Bleomycin-induced lung injury causes increased fibroblast numbers in the lung and pulmonary fibrosis. Studies of fibroblasts isolated from such injured lungs have revealed evidence of increased intrinsic proliferative capacity, but the mechanism is unknown. Telomerase catalyzes the addition of telomeric DNA repeats onto chromosomal ends, which is associated with increased cellular life span or immortality. To examine whether telomerase might play a role in regulating fibroblast proliferative capacity in pulmonary fibrosis, lung fibroblasts were isolated from rats treated with endotracheal injections of phosphate-buffered saline or bleomycin. At selected time points, the rats were killed and lung fibroblasts isolated. The isolated cells and lung tissue were then used in experiments for measurement of telomerase activity. The results show undetectable telomerase activity in fibroblasts isolated from control uninjured lungs, or in the control lung tissue extracts. Similar results were obtained in cells and lung tissue from Days 1, 3, and 28 bleomycin-injured lungs. However, significant telomerase activity was detected in fibroblasts and tissue extracts isolated from Days 7, 14, and 21 bleomycin-treated rat lungs, with maximal activity observed in the Day 14 samples. Analysis of the isolated cells for telomerase messenger RNA or reverse transcriptase expression, combined with alpha-smooth-muscle actin expression by immunohistochemistry, revealed that telomerase expression localized primarily to nonmyofibroblasts. These findings suggest that in addition to elevated growth factor expression, the injured lung fibroblast population may contain cells with increased life span, which could contribute to the observed overall increase in lung fibroblast numbers.


Assuntos
Bleomicina/efeitos adversos , Pulmão/efeitos dos fármacos , RNA , Telomerase/metabolismo , Animais , Sequência de Bases , Primers do DNA , Proteínas de Ligação a DNA , Indução Enzimática , Fibroblastos/enzimologia , Pulmão/enzimologia , Pulmão/patologia , Masculino , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Telomerase/genética
11.
J Invest Dermatol ; 114(3): 480-6, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10692106

RESUMO

Damage to human skin due to ultraviolet light from the sun (photoaging) and damage occurring as a consequence of the passage of time (chronologic or natural aging) are considered to be distinct entities. Photoaging is caused in part by damage to skin connective tissue by increased elaboration of collagen-degrading matrix metalloproteinases, and by reduced collagen synthesis. As matrix metalloproteinase levels are known to rise in fibroblasts as a function of age, and as oxidant stress is believed to underlie changes associated with both photoaging and natural aging, we determined whether natural skin aging, like photoaging, gives rise to increased matrix metalloproteinases and reduced collagen synthesis. In addition, we determined whether topical vitamin A (retinol) could stimulate new collagen deposition in sun-protected aged skin, as it does in photoaged skin. Sun-protected skin samples were obtained from 72 individuals in four age groups: 18-29 y, 30-59 y, 60-79 y, and 80+ y. Histologic and cellular markers of connective tissue abnormalities were significantly elevated in the 60-79 y and 80+ y groups, compared with the two younger age groups. Increased matrix metalloproteinase levels and decreased collagen synthesis/expression were associated with this connective tissue damage. In a separate group of 53 individuals (80+ y of age), topical application of 1% vitamin A for 7 d increased fibroblast growth and collagen synthesis, and concomitantly reduced the levels of matrix-degrading matrix metalloproteinases. Our findings indicate that naturally aged, sun-protected skin and photoaged skin share important molecular features including connective tissue damage, elevated matrix metalloproteinase levels, and reduced collagen production. In addition, vitamin A treatment reduces matrix metalloproteinase expression and stimulates collagen synthesis in naturally aged, sun-protected skin, as it does in photoaged skin.


Assuntos
Colágeno/metabolismo , Envelhecimento da Pele/efeitos dos fármacos , Pele/citologia , Vitamina A/farmacologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/fisiologia , Divisão Celular/efeitos dos fármacos , Colágeno/farmacologia , Tecido Conjuntivo/efeitos dos fármacos , Fibroblastos/citologia , Humanos , Metaloproteinases da Matriz/efeitos dos fármacos , Pessoa de Meia-Idade , Pró-Colágeno/biossíntese , Pele/química , Pele/metabolismo , Envelhecimento da Pele/fisiologia , Estimulação Química
12.
Am J Respir Cell Mol Biol ; 21(6): 658-65, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10572062

RESUMO

Fibroblast differentiation to the myofibroblast phenotype is associated with alpha-smooth-muscle actin (alpha-SMA) expression and regulated by cytokines. Among these, transforming growth factor (TGF)-beta(1) and interleukin (IL)-1beta can stimulate and inhibit myofibroblast differentiation, respectively. IL-1beta inhibits alpha-SMA expression by inducing apoptosis selectively in myofibroblasts via induction of nitric oxide synthase (inducible nitric oxide synthase [iNOS]). Because TGF-beta is known to inhibit iNOS expression, this study was undertaken to see if this cytokine can protect against IL-1beta-induced myofibroblast apoptosis. Rat lung fibroblasts were treated with IL-1beta and/or TGF-beta(1) and examined for expression of alpha-SMA, iNOS, and the apoptotic regulatory proteins bax and bcl-2. The results show that TGF-beta(1) caused a virtually complete suppression of IL-1beta-induced iNOS expression while preventing the decline in alpha-SMA expression or the myofibroblast subpopulation. TGF-beta(1) treatment also completely suppressed the IL-1beta-induced apoptosis in myofibroblasts. IL-1beta-induced apoptosis was associated with a significant decline in expression of the antiapoptotic protein bcl-2, which was prevented by concomitant TGF-beta(1) treatment. The level of the proapoptotic protein bax, however, was not significantly altered by either cytokine. These data suggest that TGF-beta(1) inhibits IL-1beta-induced apoptosis in myofibroblasts by at least two mechanisms, namely, the suppression of iNOS expression and the prevention of a decline in bcl-2 expression. Thus, TGF-beta(1) may be additionally important in fibrosis by virtue of this novel ability to promote myofibroblast survival by preventing the myofibroblast from undergoing apoptosis.


Assuntos
Actinas/metabolismo , Apoptose/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Interleucina-1/farmacologia , Pulmão/efeitos dos fármacos , Músculo Liso/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Ratos , Proteína X Associada a bcl-2
14.
Int J Mol Med ; 1(1): 43-53, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9852197

RESUMO

Pulmonary fibrosis is commonly characterized by inflammation of the alveolar wall, leading to derangement of normal alveolar architecture, and interstitial as well as intra-alveolar fibrosis. The process involves cellular interactions via a complex cytokine network and heightened collagen gene expression with abnormal deposition in the lung. Recent studies have identified a myriad of cytokines with potential roles in pulmonary fibrosis. Based on in vivo antibody neutralization studies, important roles for tumor necrosis á (TNFá), macrophage inflammatory protein 1á (MIP-1á) and transforming growth factor (TGF), have been established. The recent demonstration that the eosinophil is a major source for several of these key pro-fibrogenic cytokines during the early stages of fibrosis, strongly suggest a role for the eosinophil in pulmonary fibrosis. In vitro, eosinophils can elaborate factors capable of stimulating fibroblast proliferation, and their presence in lungs undergoing many forms of pulmonary fibrosis has been well documented. Further support for a role for eosinophils in pulmonary fibrosis are suggested by clinical data showing a correlation between lung eosinophil count and a poor prognosis and decreased responsiveness to therapy. This review will focus on the recent findings, which suggest novel potential roles for the eosinophil in the pathogenesis of pulmonary fibrosis.


Assuntos
Eosinófilos/fisiologia , Fibrose Pulmonar/etiologia , Animais , Quimiotaxia de Leucócito , Citocinas/imunologia , Modelos Animais de Doenças , Eosinófilos/imunologia , Humanos , Fibrose Pulmonar/imunologia
15.
J Leukoc Biol ; 64(5): 657-66, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9823772

RESUMO

Eosinophils are known to express cytokines capable of promoting fibrosis. Interleukin-5 (IL-5) is important in regulating eosinophilopoiesis, eosinophil recruitment and activation. Lung IL-5 expression is elevated in pulmonary fibrosis, wherein the eosinophil is a primary source of fibrogenic cytokines. To determine the role of IL-5 in pulmonary fibrosis, the effects of anti-IL-5 antibody were investigated in a model of bleomycin-induced pulmonary fibrosis. Fibrosis was induced in mice by endotracheal bleomycin treatment. Animals were also treated with either anti-IL-5 antibody or control IgG. Lungs were then analyzed for fibrosis, eosinophil influx, chemotactic activity, and cytokine expression. The results show that a primary chemotactic activity at the height of eosinophil recruitment is IL-5. Furthermore, anti-IL-5 antibody caused significant reduction in lung eosinophilia, cytokine expression, and fibrosis. These findings taken together suggest an important role for IL-5 in pulmonary fibrosis via its ability to regulate eosinophilic inflammation, and thus eosinophil-dependent fibrogenic cytokine production.


Assuntos
Bleomicina/efeitos adversos , Eosinófilos/fisiologia , Interleucina-5/fisiologia , Fibrose Pulmonar/induzido quimicamente , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Eosinófilos/metabolismo , Feminino , Interleucina-5/antagonistas & inibidores , Interleucina-5/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos CBA , Fibrose Pulmonar/fisiopatologia , Fibrose Pulmonar/prevenção & controle , Organismos Livres de Patógenos Específicos
16.
J Leukoc Biol ; 64(4): 528-36, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9766634

RESUMO

Previously, macrophage inflammatory protein-1alpha (MIP-1alpha), a member of the C-C chemokine family, has been implicated in bleomycin-induced pulmonary fibrosis, a model of the human disease idiopathic pulmonary fibrosis. Neutralization of MIP-1alpha protein with anti-MIP-1alpha antibodies significantly attenuated both mononuclear phagocyte recruitment and pulmonary fibrosis in bleomycin-challenged CBA/J mice. However, the specific stimuli for MIP-1alpha expression in the bleomycin-induced lesion have not been characterized. In this report, two mediators of the inflammatory response to bleomycin, tumor necrosis factor (TNF) and interleukin-6 (IL-6), were evaluated as putative stimuli for MIP-1alpha expression after bleomycin challenge in CBA/J mice. Elevated levels of bioactive TNF and IL-6 were detected in bronchoalveolar lavage (BAL) fluid and lung homogenates from bleomycin-treated CBA/J mice at time points post-bleomycin challenge, which precede MIP-1alpha protein expression. Treatment of bleomycin-challenged mice with soluble TNF receptor (sTNFr) or anti-IL-6 antibodies significantly decreased MIP-1alpha protein expression in the lungs. Furthermore, normal alveolar macrophages secreted elevated levels of MIP-1alpha protein in response to treatment with TNF plus IL-6 or bleomycin plus IL-6, but not TNF, bleomycin, or IL-6 alone. Finally, leukocytes recovered from the BAL fluid of bleomycin-challenged mice secreted higher levels of MIP-1alpha protein, compared to controls, when treated with TNF alone. Based on the data presented here, we propose that TNF and IL-6 are part of a cytokine network that modulates MIP-1alpha protein expression in the profibrotic inflammatory lesion during the response to intratracheal bleomycin challenge.


Assuntos
Bleomicina/toxicidade , Citocinas/fisiologia , Interleucina-6/fisiologia , Pulmão/fisiopatologia , Proteínas Inflamatórias de Macrófagos/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos/farmacologia , Líquido da Lavagem Broncoalveolar/imunologia , Quimiocina CCL3 , Quimiocina CCL4 , Humanos , Imunoterapia , Hibridização In Situ , Interleucina-6/análise , Interleucina-6/biossíntese , Pulmão/efeitos dos fármacos , Pulmão/patologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Camundongos , Camundongos Endogâmicos CBA , Modelos Biológicos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/fisiopatologia , RNA Mensageiro/biossíntese , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/uso terapêutico , Fatores de Tempo , Transcrição Gênica , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/biossíntese
17.
Am J Respir Cell Mol Biol ; 16(4): 438-47, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9115755

RESUMO

Eosinophils are primary sources of fibrogenic cytokines in lung fibrosis, and interleukin (IL)-5 is important in their differentiation, proliferation, recruitment and activation. To investigate the potential role of this cytokine, lung IL-5 expression was examined in a murine model of bleomycin-induced pulmonary fibrosis. Analysis of lung RNA showed significant increases in lung IL-5 mRNA content between days 3 and 14 after induction of lung injury, which decreased toward control levels after day 21. In situ hybridization revealed essentially no detectable IL-5 mRNA expression before day 3, but showed elevated expression in mononuclear cells and eosinophils between days 3 and 14, localized within areas of active fibrosis. After 21 days, the intensity and number of IL-5-expressing cells significantly declined. Immunostaining with anti-IL-5 antibodies confirmed the predominant IL-5 expression by mononuclear cells and eosinophils in areas of active fibrosis. The kinetics of increase in the number of cells expressing significant IL-5 mRNA in lung sections paralleled that for IL-5 mRNA expression in whole-lung homogenates. These results demonstrate for the first time that IL-5 is upregulated in this murine model and suggest a novel role for this cytokine in pulmonary fibrosis via its ability to recruit and activate eosinophils.


Assuntos
Bleomicina/efeitos adversos , Interleucina-5/genética , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Northern Blotting , Feminino , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos CBA , Reação em Cadeia da Polimerase , Fibrose Pulmonar/induzido quimicamente , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
18.
J Vasc Surg ; 25(3): 446-52, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9081125

RESUMO

PURPOSE: This investigation was designed to test the hypothesis that transforming growth factor-beta 1 (TGF-beta 1) regulates lysyl oxidase secretion from vascular smooth muscle cells. Lysyl oxidase is an enzyme that catalyzes an essential step in collagen and elastin cross-linking in the extracellular matrix, and TGF-beta 1 has been implicated in the pathogenesis of restenosis after vascular injury. The effect of TGF-beta 1 on lysyl oxidase in vascular smooth muscle cells has not been previously defined. METHODS: Rat aortic smooth muscle cells were grown in culture to confluence. Cells in passage 2 to 6 were incubated for 24 hours in media containing 0.1, 0.5, 1.0, or 10.0 ng/ml of TGF-beta 1. Lysyl oxidase activity in the media was quantitated with a tritium-release bioassay against an insoluble 3H-labeled aortic clastin substrate. Northern blot analyses were performed to determine steady-state levels of lysyl oxidase mRNA in the smooth muscle cells. RESULTS: Lysyl oxidase activity in the media increased 1.5-fold above control levels after exposure to 10 ng/ml of TGF-beta 1 (p < 0.01). This increase in lysyl oxidase activity was associated with a concentration-dependent increase in steady-state levels of lysyl oxidase mRNA, being 4.3- and 6.2-fold above control levels after exposure to 1 and 10 ng/ml TGF-beta 1, respectively (p < 0.01). The observed increase in steady-state lysyl oxidase mRNA after exposure to TGF-beta 1 was also time-dependent over the 24-hour experimental period. CONCLUSIONS: TGF-beta 1 appears to regulate lysyl oxidase in cultured rat aortic smooth muscle cells. Increases in lysyl oxidase activity may be one of the mechanisms by which TGF-beta 1 contributes to arterial restenosis after vascular injury.


Assuntos
Músculo Liso Vascular/enzimologia , Proteína-Lisina 6-Oxidase/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Aorta Torácica/citologia , Aorta Torácica/enzimologia , Células Cultivadas , Relação Dose-Resposta a Droga , Masculino , Proteína-Lisina 6-Oxidase/genética , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/fisiologia
19.
J Immunol ; 158(3): 1392-9, 1997 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9013984

RESUMO

IL-1 is important in regulating lung inflammation and potentially fibrosis as well. To clarify the role of this cytokine vis-à-vis changes in lung fibroblast phenotype in pulmonary fibrosis, the effects of IL-1beta on isolated lung fibroblasts were examined. Rat lung fibroblasts were treated with increasing doses of IL-1beta and examined for effects on cell number, alpha-smooth muscle actin expression, apoptosis, nitric oxide (NO) production, and contractility in collagen gels. The results show that IL-1beta caused dose-dependent down-regulation of alpha-smooth muscle actin protein and mRNA expression. The kinetics of mRNA inhibition was rapid and preceded the effects on protein expression. This IL-1beta-induced decrease in actin expression was associated with inhibition of contractility evaluated using fibroblast-populated collagen gels. Since IL-1beta inhibition of actin expression was accompanied by reduction in cell number, the effect on apoptosis was examined. Significant increase in the number of apoptotic nuclei and DNA fragmentation was observed upon IL-1beta treatment, with a dose-response curve that mirrored that for the decline in actin-positive cells. More than one-half of the apoptotic cells were actin positive at high IL-1beta doses, suggesting that the actin-expressing cells may be more susceptible to IL-1beta-induced apoptosis. IL-1beta also induced NO production in these cells, which was inhibited by NG-monomethyl-L-arginine. Similarly, IL-1beta-induced apoptosis and inhibition of actin expression were inhibited by this arginine analogue. Hence, induction of apoptosis by IL-1beta via NO production may be an important mechanism for regulating lung fibroblast alpha-smooth muscle actin expression, and consequently its contractile phenotype as well.


Assuntos
Actinas/metabolismo , Interleucina-1/farmacologia , Pulmão/citologia , Animais , Apoptose , Células Cultivadas , Colágeno/química , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Géis , Expressão Gênica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Endogâmicos F344 , ômega-N-Metilarginina/farmacologia
20.
J Immunol ; 158(2): 954-9, 1997 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-8993016

RESUMO

Despite abundant evidence documenting the importance of TNF-alpha in the pathogenesis of pulmonary fibrosis, its actual role has not been fully elucidated. Recent observations also indicate that eosinophils found in fibrotic lung express elevated levels of cytokines known to be important in lung fibrosis. These findings suggest a possible role for TNF-alpha in eosinophil recruitment and cytokine expression in this disease. To examine this hypothesis, pulmonary fibrosis was induced in mice by endotracheal bleomycin treatment, and separate groups of animals were also treated with either anti-TNF-alpha Ab or control serum. On days 7 and 14 post-bleomycin treatment, lungs were harvested and analyzed for fibrosis, cytokine expression, and eosinophil influx. Anti-TNF-alpha caused a significant reduction in lung fibrosis, as indicated by a reduction in hydroxyproline content, which was accompanied by suppression of lung TGF-beta1, IL-5, and JE mRNA expression. Examination of tissue sections revealed a significant reduction in lung eosinophils and overall cellularity by anti-TNF-alpha treatment without a significant effect on the number of lung macrophages. The number of IL-5-expressing cells was also significantly reduced by anti-TNF-alpha treatment. Since IL-5 is important in eosinophil differentiation, activation, and recruitment, these findings suggest a novel mechanism by which TNF-alpha could mediate pulmonary fibrosis via induction of IL-5-mediated eosinophil recruitment and fibrogenic cytokine production. Since these eosinophil-derived cytokines include JE/monocyte chemotactic factor-1 and TGF-beta1, this cytokine networking orchestrated by TNF-alpha could, in turn, amplify the inflammatory response and drive the progression to fibrosis and end-stage lung disease.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Eosinófilos/efeitos dos fármacos , Fibrose Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Bleomicina/administração & dosagem , Bleomicina/toxicidade , Quimiocina CCL2/biossíntese , Quimiocina CCL2/metabolismo , Citocinas/biossíntese , Eosinófilos/metabolismo , Feminino , Interleucina-5/biossíntese , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Fibrose Pulmonar/etiologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...