Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
iScience ; 26(5): 106570, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-37250334

RESUMO

Cell reprogramming to a myofibroblast responsible for the pathological accumulation of extracellular matrix is fundamental to the onset of fibrosis. Here, we explored how condensed chromatin structure marked by H3K72me3 becomes modified to allow for activation of repressed genes to drive emergence of myofibroblasts. In the early stages of myofibroblast precursor cell differentiation, we discovered that H3K27me3 demethylase enzymes UTX/KDM6B creates a delay in the accumulation of H3K27me3 on nascent DNA revealing a period of decondensed chromatin structure. This period of decondensed nascent chromatin structure allows for binding of pro-fibrotic transcription factor, Myocardin-related transcription factor A (MRTF-A) to nascent DNA. Inhibition of UTX/KDM6B enzymatic activity condenses chromatin structure, prevents MRTF-A binding, blocks activation of the pro-fibrotic transcriptome, and results in an inhibition of fibrosis in lens and lung fibrosis models. Our work reveals UTX/KDM6B as central coordinators of fibrosis, highlighting the potential to target its demethylase activity to prevent organ fibrosis.

2.
Biomolecules ; 13(5)2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37238610

RESUMO

Junctional epidermolysis bullosa (JEB) patients experience skin and epithelial fragility due to a pathological deficiency in genes associated with epidermal adhesion. Disease severity ranges from post-natal lethality to localized skin involvement with persistent blistering followed by granulation tissue formation and atrophic scarring. We evaluated the potential of utilizing Trametinib, an MEK inhibitor previously shown to target fibrosis, with and without the documented EB-anti-fibrotic Losartan for reducing disease severity in a mouse model of JEB; Lamc2jeb mice. We found that Trametinib treatment accelerated disease onset and decreased epidermal thickness, which was in large part ameliorated by Losartan treatment. Interestingly, a range of disease severity was observed in Trametinib-treated animals that tracked with epidermal thickness; those animals grouped with higher disease severity had thinner epidermis. To examine if the difference in severity was related to inflammation, we conducted immunohistochemistry for the immune cell markers CD3, CD4, CD8, and CD45 as well as the fibrotic marker αSMA in mouse ears. We used a positive pixel algorithm to analyze the resulting images and demonstrated that Trametinib caused a non-significant reduction in CD4 expression that inversely tracked with increased fibrotic severity. With the addition of Losartan to Trametinib, CD4 expression was similar to control. Together, these data suggest that Trametinib causes a reduction in both epidermal proliferation and immune cell infiltration/proliferation, with concurrent acceleration of skin fragility, while Losartan counteracts Trametinib's adverse effects in a mouse model of JEB.


Assuntos
Epidermólise Bolhosa Juncional , Camundongos , Animais , Epidermólise Bolhosa Juncional/genética , Epidermólise Bolhosa Juncional/patologia , Losartan , Pele/patologia , Epiderme
3.
Connect Tissue Res ; 63(3): 200-209, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35321605

RESUMO

This biography of Dr. Joel Rosenbloom is published on the occasion of the 50th anniversary of the journal. Dr. Rosenbloom presents the scientific milestones and achievements throughout his career emphasizing events that have spurred him to launch into a career in biomedical research and education. The biography spans several decades of the life and achievements of a distinguished physician scientist whose dedication to science demonstrates the development of new insights into a variety of connective tissues through technological advances and insightful approaches.


Assuntos
Pesquisa Biomédica , Pesquisa Biomédica/educação , Humanos , Masculino
4.
Endocrinol Diabetes Metab ; 4(2): e00195, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33855203

RESUMO

Introduction: Fibrosis is characterized by dysregulation and accumulation of extracellular matrix. Peyronie's disease and Dupuytren disease are fibroproliferative disorders of the tunica albuginea of the penis and fascia of the hand, respectively. Chronic hyperglycaemia due to diabetes mellitus can also lead to tissue injury and fibrosis. A meta-analysis has shown a relationship between Dupuytren disease and diabetes (overall odds ratio, 3.1; 95% confidence interval, 2.7-3.5). This review explores commonalities in the pathogenesis of Peyronie's disease, Dupuytren disease and diabetes. Methods: A search of the PubMed database was conducted using the search terms "diabetes" AND "Peyronie's disease"; and "diabetes" AND "Dupuytren." Results: Genome-wide association and gene expression studies conducted with tissue from people with Peyronie's disease or Dupuytren disease identified signalling pathways associated with wingless-type mammary-tumour virus integration site signalling, extracellular matrix modulation and inflammation. Biochemical studies confirmed the importance of these pathways in the pathogenesis of fibrosis with Peyronie's disease and Dupuytren disease. Dysregulation of matrix metalloproteinase activity associated with extracellular matrix breakdown was implicated in fibroproliferative complications of diabetes and in the aetiology of Peyronie's disease and Dupuytren disease. A notable percentage of people with diabetes have comorbid Peyronie's disease and/or Dupuytren disease. Conclusions: Studies have not been performed to identify fibroproliferative pathways that all 3 conditions might have in common, but data suggest that common pathways are involved in the fibroproliferative processes of Peyronie's disease, Dupuytren disease, and diabetes.


Assuntos
Diabetes Mellitus/etiologia , Contratura de Dupuytren/etiologia , Estudo de Associação Genômica Ampla , Induração Peniana/etiologia , Doença Crônica , Complicações do Diabetes/complicações , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Contratura de Dupuytren/genética , Contratura de Dupuytren/patologia , Matriz Extracelular/metabolismo , Fáscia/patologia , Feminino , Fibrose , Mãos , Humanos , Hiperglicemia/etiologia , Masculino , Metaloproteinases da Matriz/metabolismo , Induração Peniana/genética , Induração Peniana/patologia , Pênis/patologia , Transdução de Sinais
5.
Exp Dermatol ; 30(1): 132-145, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33211348

RESUMO

Keloid disorder, a group of fibroproliferative skin diseases, is characterized by unremitting accumulation of the extracellular matrix (ECM) of connective tissue, primarily collagen, to develop cutaneous tumors on the predilection sites of skin. There is a strong genetic predisposition for keloid formation, and individuals of African and Asian ancestry are particularly prone. The principal cell type responsible for ECM accumulation is the myofibroblast derived from quiescent resident skin fibroblasts either through trans-differentiation or from keloid progenitor stem cells with capacity for multi-lineage differentiation and self-renewal. The biosynthetic pathways leading to ECM accumulation are activated by several cytokines, but particularly by TGF-ß signalling. The mechanical properties of the cellular microenvironment also play a critical role in the cell's response to TGF-ß, as demonstrated by culturing of fibroblasts derived from keloids and control skin on substrata with different degrees of stiffness. These studies also demonstrated that culturing of fibroblasts on tissue culture plastic in vitro does not reflect their biosynthetic capacity in vivo. Collectively, our current understanding of the pathogenesis of keloids suggests a complex network of interacting cellular, molecular and mechanical factors, with distinct pathways leading to myofibroblast differentiation and activation. Keloids can serve as a model system of fibrotic diseases, a group of currently intractable disorders, and deciphering of the critical pathogenetic steps leading to ECM accumulation is expected to identify targets for pharmacologic intervention, not only for keloids but also for a number of other, both genetic and acquired, fibrotic diseases.


Assuntos
Matriz Extracelular , Fibroblastos/metabolismo , Queloide/genética , Queloide/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Transdiferenciação Celular , Fibroblastos/fisiologia , Fibronectinas/genética , Fibronectinas/metabolismo , Expressão Gênica , Humanos , Queloide/patologia , Miofibroblastos , Transdução de Sinais , Células-Tronco , Transcriptoma
6.
Nat Rev Rheumatol ; 14(7): 421-432, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29789665

RESUMO

The discovery and validation of biomarkers resulting from technological advances in the analysis of genomic, transcriptomic, lipidomic and metabolomic pathways involved in the pathogenesis of complex human diseases have led to the development of personalized and rationally designed approaches for the clinical management of such disorders. Although some of these approaches have been applied to systemic sclerosis (SSc), an unmet need remains for validated, non-invasive biomarkers to aid in the diagnosis of SSc, as well as in the assessment of disease progression and response to therapeutic interventions. Advances in global transcriptomic technology over the past 15 years have enabled the assessment of microRNAs that circulate in the blood of patients and the analysis of the macromolecular content of a diverse group of lipid bilayer membrane-enclosed extracellular vesicles, such as exosomes and other microvesicles, which are released by all cells into the extracellular space and circulation. Such advances have provided new opportunities for the discovery of biomarkers in SSc that could potentially be used to improve the design and evaluation of clinical trials and that will undoubtedly enable the development of personalized and individualized medicine for patients with SSc.


Assuntos
Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Escleroderma Sistêmico/diagnóstico , Biomarcadores/metabolismo , Ensaios Clínicos como Assunto , Progressão da Doença , Diagnóstico Precoce , Humanos , Medicina de Precisão , Escleroderma Sistêmico/sangue , Escleroderma Sistêmico/tratamento farmacológico , Resultado do Tratamento
7.
J Surg Res ; 227: 198-210, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29804854

RESUMO

BACKGROUND: Intra-abdominal adhesions are a major cause of morbidity after abdominal or gynecologic surgery. However, knowledge about the pathogenic mechanism(s) is limited, and there are no effective treatments. Here, we investigated a mouse model of bowel adhesion formation and the effect(s) of an Federal Drug Administration-approved drug (trametinib) in preventing adhesion formation. MATERIALS AND METHODS: C57BL/6 mice were used to develop a consistent model of intra-abdominal adhesion formation by gentle cecal abrasion with mortality rates of <10%. Adhesion formation was analyzed histologically and immunochemically to characterize the expression of pro-fibrotic marker proteins seen in pathologic scaring and included alpha smooth muscle actin (αSMA) and fibronectin EDA (FNEDA) which arises from alternative splicing of the fibronectin messenger RNA resulting in different protein isoforms. Trichrome staining assessed collagen deposition. Quantitative polymerase chain reaction analysis of RNA isolated from adhesions by laser capture microscopy was carried out to assess pro-fibrotic gene expression. To block adhesion formation, trametinib was administered via a subcutaneous osmotic pump. RESULTS: Adhesions were seen as early as post-operative day 1 with extensive adhesions being formed and vascularized by day 5. The expression of the FNEDA isoform occurred first with subsequent expression of αSMA and collagen. The drug trametinib was chosen for in vivo studies because it effectively blocked the mesothelial to mesenchymal transition of rat mesothelium. Trametinib, at the highest dose used (3 mg/kg/d), prevented adhesion formation while at lower doses, adhesions were usually limited, as evidenced by the presence of FNEDA isoform but not αSMA. CONCLUSIONS: Cecal abrasion in mice is a reliable model to study abdominal adhesions, which can be ameliorated using the MEK1/2 inhibitor trametinib. While blocking adhesion formation at the cell and molecular levels, trametinib, at the therapeutic doses utilized, did not impair the wound healing at the laparotomy site.


Assuntos
Ceco/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Complicações Pós-Operatórias/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Piridonas/farmacologia , Pirimidinonas/farmacologia , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Parede Abdominal/cirurgia , Animais , Ceco/efeitos dos fármacos , Ceco/cirurgia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peritônio/efeitos dos fármacos , Peritônio/patologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Aderências Teciduais/etiologia , Aderências Teciduais/patologia , Aderências Teciduais/prevenção & controle , Cicatrização/efeitos dos fármacos
8.
J Cancer Treatment Diagn ; 2(4): 7-9, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30949622

RESUMO

Radiation induced fibrosis (RIF) is a common morbidity in patients being treated for cancer with radiation. Off-target effects result in intense inflammatory responses which ultimately results in the generation of extracellular matrix (ECM) producing myofibroblasts which mediate a progressive fibrosis resulting in scarring and organ and tissue dysfunction. Unfortunately, currently, there are no effective therapies to block the excess accumulation of ECM. We have previously reported on the use of trametinib, a MEK inhibitor, to essentially block the formation of abdominal adhesions in a mouse model of cecal abrasion. Using this drug in the mouse model, the complete trans-differentiation of precursor cells into ECM-producing myofibroblasts was blocked. Trametinib is a potentially powerful drug to thwart organ and tissue fibrosis in RIF because it has a potential dual function in that it may block RIF as well as prevent radiation-resistance. Given the intractability of RIF, trametinib should be considered for more extensive testing.

9.
Methods Mol Biol ; 1627: 1-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836191

RESUMO

Human fibrotic diseases constitute a major health problem worldwide owing to the large number of affected individuals, the incomplete knowledge of the fibrotic process pathogenesis, the marked heterogeneity in their etiology and clinical manifestations, the absence of appropriate and fully validated biomarkers, and, most importantly, the current void of effective disease-modifying therapeutic agents. The fibrotic disorders encompass a wide spectrum of clinical entities including systemic fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, and nephrogenic systemic fibrosis, as well as numerous organ-specific disorders including radiation-induced fibrosis and cardiac, pulmonary, liver, and kidney fibrosis. Although their causative mechanisms are quite diverse and in several instances have remained elusive, these diseases share the common feature of an uncontrolled and progressive accumulation of fibrotic tissue in affected organs causing their dysfunction and ultimate failure. Despite the remarkable heterogeneity in the etiologic mechanisms responsible for the development of fibrotic diseases and in their clinical manifestations, numerous studies have identified activated myofibroblasts as the common cellular element ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. Critical signaling cascades, initiated primarily by transforming growth factor-ß (TGF-ß), but also involving numerous cytokines and signaling molecules which stimulate profibrotic reactions in myofibroblasts, offer potential therapeutic targets. Here, we briefly review the current knowledge of the molecular mechanisms involved in the development of tissue fibrosis and point out some of the most important challenges to research in the fibrotic diseases and to the development of effective therapeutic approaches for this often fatal group of disorders. Efforts to further clarify the complex pathogenetic mechanisms of the fibrotic process should be encouraged to attain the elusive goal of developing effective therapies for these serious, untreatable, and often fatal disorders.


Assuntos
Fibrose/etiologia , Fibrose/metabolismo , Animais , Biomarcadores , Proteínas Morfogenéticas Ósseas/metabolismo , Suscetibilidade a Doenças , Matriz Extracelular/metabolismo , Fibrose/diagnóstico , Fibrose/terapia , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Terapia de Alvo Molecular , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Estresse Oxidativo , Pesquisa , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
10.
Matrix Biol ; 51: 37-46, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26844756

RESUMO

Keloids, fibroproliferative dermal tumors with effusive accumulation of extracellular matrix (ECM) components, particularly collagen, result from excessive expression of growth factors and cytokines. The etiology of keloids is unknown but they occur after dermal injury in genetically susceptible individuals, and they cause both physical and psychological distress for the affected individuals. Several treatment methods for keloids exist, including the combination therapy of surgical excision followed by intralesional steroid therapy, however, they have high recurrence rate regardless of the current treatment method. Improved understanding of the pathomechanisms leading to keloid formation will hopefully identify pathways that serve as specific targets to improve therapy for this devastating, currently intractable, disorder.


Assuntos
Fibrose/tratamento farmacológico , Queloide/tratamento farmacológico , Pele/patologia , Esteroides/uso terapêutico , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibroblastos/patologia , Fibrose/patologia , Fibrose/cirurgia , Humanos , Queloide/patologia , Queloide/cirurgia , Pele/lesões , Cicatrização/efeitos dos fármacos
11.
Matrix Biol ; 51: 47-54, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26827712

RESUMO

Animal models are crucial for the study of fibrosis. Keloids represent a unique type of fibrotic scarring that occurs only in humans, thus presenting a challenge for those studying the pathogenesis of this disease and its therapeutic options. Here, several animal models of fibrosis currently in use are described, emphasizing recent progress and highlighting encouraging challenges.


Assuntos
Cicatriz Hipertrófica/genética , Fibrose/genética , Queloide/genética , Animais , Cicatriz Hipertrófica/fisiopatologia , Cicatriz Hipertrófica/terapia , Modelos Animais de Doenças , Fibroblastos/patologia , Fibrose/fisiopatologia , Fibrose/terapia , Humanos , Queloide/fisiopatologia , Queloide/terapia , Camundongos , Proteínas Serina-Treonina Quinases/genética
12.
Matrix Biol ; 51: 55-64, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26825317

RESUMO

Peritoneal adhesions, primarily caused by surgical procedures, are the leading cause of pelvic pain, bowel obstruction, and infertility. TGF-ß1 and IL-6 have been found to be elevated in the peritoneal fluid of patients during/after abdominal surgery. However, it remains to be determined whether these cytokines interact and facilitate adhesion formation by promoting mesothelial to mesenchymal transition (MMT). In the present study, isolated rat peritoneal mesothelial cells were treated with TGF-ß1 and/or IL-6 which elicited MMT as determined by morphologic and biochemical techniques. During this transition, cellular morphology changed from that of cobblestone polygonal cells to elongated/spindle-shaped fibroblast-like cells. There was decreased expression of genes characteristic of mesothelial cells, such as E-cadherin, and increased expression of genes characteristic of the myofibroblast phenotype, including α-smooth muscle actin and the EDA form of fibronectin, both of which appear to mediate the transfer of force to the extracellular matrix. Partial characterization of relevant signaling pathways identified Erk1/2 activation, which was enhanced by combined TGF-ß1/IL-6 administration, as a crucial necessary factor in the transition. Erk1/2 activation as well as the phosphorylation of the linker region of Smad2 and MMT could be blocked by the MEK inhibitor, U0126, suggesting that such activation may be a potential pharmaceutical target to prevent MMT. In addition, the phenotypic transition could be prevented by hydrocortisone.


Assuntos
Transição Epitelial-Mesenquimal/genética , Interleucina-6/administração & dosagem , Peritônio/metabolismo , Fator de Crescimento Transformador beta1/administração & dosagem , Animais , Líquido Ascítico/metabolismo , Líquido Ascítico/patologia , Butadienos/administração & dosagem , Caderinas/biossíntese , Fibronectinas/biossíntese , Humanos , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Nitrilas/administração & dosagem , Peritônio/patologia , Fosforilação , Ratos , Proteína Smad2/biossíntese , Fator de Crescimento Transformador beta1/metabolismo
13.
Matrix Biol ; 51: 14-25, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26807756

RESUMO

Fibrotic diseases constitute a world-wide major health problem, but research support remains inadequate in comparison to the need. Although considerable understanding of the pathogenesis of fibrotic reactions has been attained, no completely effective therapies exist. Although fibrotic disorders are diverse, it is universally appreciated that a particular cell type with unique characteristics, the myofibroblast, is responsible for replacement of functioning tissue with non-functional scar tissue. Understanding the cellular and molecular mechanisms responsible for the creation of myofibroblasts and their activities is central to the development of therapies. Critical signaling cascades, initiated primarily by TGF-ß, but also involving other cytokines which stimulate pro-fibrotic reactions in the myofibroblast, offer potential therapeutic targets. However, because of the multiplicity and complex interactions of these signaling pathways, it is very unlikely that any single drug will be successful in modifying a major fibrotic disease. Therefore, we have chosen to examine the effectiveness of administration of several drug combinations in a mouse pneumoconiosis model. Such treatment proved to be effective. Because fibrotic diseases that tend to be chronic, are difficult to monitor, and are patient variable, implementation of clinical trials is difficult and expensive. Therefore, we have made efforts to identify and validate non-invasive biomarkers found in urine and blood. We describe the potential utility of five such markers: (i) the EDA form of fibronectin (Fn(EDA)), (ii) lysyl oxidase (LOX), (iii) lysyl oxidase-like protein 2 (LoxL2), (iv) connective tissue growth factor (CTGF, CCNII), and (v) the N-terminal propeptide of type III procollagen (PIIINP).


Assuntos
Biomarcadores/sangue , Biomarcadores/urina , Pneumoconiose/sangue , Pneumoconiose/urina , Aminoácido Oxirredutases/sangue , Aminoácido Oxirredutases/urina , Animais , Fator de Crescimento do Tecido Conjuntivo/sangue , Fator de Crescimento do Tecido Conjuntivo/urina , Modelos Animais de Doenças , Fibronectinas/sangue , Fibronectinas/urina , Humanos , Camundongos , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/urina , Pneumoconiose/patologia , Pró-Colágeno/sangue , Pró-Colágeno/urina , Receptores Depuradores Classe E/sangue
15.
Biochim Biophys Acta ; 1832(7): 1088-103, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23266403

RESUMO

The fibrotic diseases encompass a wide spectrum of entities including such multisystemic diseases as systemic sclerosis, nephrogenic systemic fibrosis and sclerodermatous graft versus host disease, as well as organ-specific disorders such as pulmonary, liver, and kidney fibrosis. Collectively, given the wide variety of affected organs, the chronic nature of the fibrotic processes, and the large number of individuals suffering their devastating effects, these diseases pose one of the most serious health problems in current medicine and a serious economic burden to society. Despite these considerations there is currently no accepted effective treatment. However, remarkable progress has been achieved in the elucidation of their pathogenesis including the identification of the critical role of myofibroblasts and the determination of molecular mechanisms that result in the transcriptional activation of the genes responsible for the fibrotic process. Here we review the origin of the myofibroblast and discuss the crucial regulatory pathways involving multiple growth factors and cytokines that participate in the pathogenesis of the fibrotic process. Potentially effective therapeutic strategies based upon this new information are considered in detail and the major challenges that remain and their possible solutions are presented. It is expected that translational efforts devoted to convert this new knowledge into novel and effective anti-fibrotic drugs will be forthcoming in the near future. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.


Assuntos
Fibrose , Miofibroblastos , Citocinas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Nefropatias/metabolismo
16.
Ann Intern Med ; 152(3): 159-66, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20124232

RESUMO

Abnormal and exaggerated deposition of extracellular matrix is the hallmark of many fibrotic diseases, including systemic sclerosis and pulmonary, liver, and kidney fibrosis. The spectrum of affected organs, the usually progressive nature of the fibrotic process, the large number of affected persons, and the absence of effective treatment pose an enormous challenge when treating fibrotic diseases. Delineation of the central role of transforming growth factor-beta (TGF-beta) and identification of the specific cellular receptors, kinases, and other mediators involved in the fibrotic process have provided a sound basis for development of effective therapies. The inhibition of signaling pathways activated by TGF-beta represents a novel therapeutic approach for the fibrotic disorders. One of these TGF-beta pathways results in the activation of the nonreceptor tyrosine kinase cellular Abelson (c-Abl), and c-Abl inhibitors, including imatinib mesylate, diminishing the fibrogenic effects of TGF-beta. Thus, recently acquired basic knowledge about the pathogenesis of the fibrotic process has enabled the development of novel therapeutic agents capable of modifying the deleterious effects of the fibrotic diseases.


Assuntos
Fibrose/tratamento farmacológico , Fibrose/etiologia , Fibrose/patologia , Humanos , Linfotoxina-alfa/fisiologia
19.
Amyloid ; 13(2): 78-85, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16911961

RESUMO

Medin amyloid is found in the medial layer of the aorta in almost 100% of the Caucasian population over 50 years of age. The medin fragment is 5.5 kDa and derives from the C2-like domain of the precursor protein lactadherin. We have previously reported immunohistochemical findings showing that medin amyloid co-localizes with elastic fibers of arteries and herein we show that lactadherin also is associated with elastic structures of human aortic material. In addition, results from in vitro binding assays demonstrate that both medin and lactadherin bind to tropoelastin in a concentration-dependent fashion, suggesting that the lactadherin-tropoelastin interaction is mediated via the medin domain. It is possible that lactadherin, which is a cell adhesion protein, in this way connects smooth muscle cells to the elastic fibers of arteries. Given that both medin and lactadherin interact with elastic fibers, elastin is probably an important component in the formation of medin amyloid.


Assuntos
Amiloide/metabolismo , Antígenos de Superfície/metabolismo , Proteínas do Leite/metabolismo , Miócitos de Músculo Liso/metabolismo , Tropoelastina/metabolismo , Amiloide/química , Antígenos de Superfície/química , Aorta/química , Aorta/metabolismo , Adesão Celular , Linhagem Celular , Humanos , Proteínas do Leite/química , Miócitos de Músculo Liso/química , Ligação Proteica , Estrutura Terciária de Proteína , Tropoelastina/química
20.
Cell Stress Chaperones ; 10(4): 285-95, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16333983

RESUMO

AFKBP65 (65-kDa FK506-binding protein) is an endoplasmic reticulum (ER)-localized peptidyl-prolyl cis-trans isomerase predicted to play a role in the folding and trafficking of secretory proteins. In previous studies, we have shown that FKBP65 is developmentally regulated and associates with the extracellular matrix protein, tropoelastin, during its maturation and transport through the ER. In this study, we show that FKBP65 is expressed in the lung with the same developmental pattern as tropoelastin and other matrix proteins. To test the hypothesis that FKBP65 is upregulated at times when extracellular matrix proteins are being actively synthesized and assembled, adult mice were treated with bleomycin to cause reinitiation of matrix protein production during the ensuing development of pulmonary fibrosis. After bleomycin instillation, FKBP65 expression was reactivated in the lung with a pattern similar to that observed for tropoelastin and type I collagen. Using human lung fibroblast cultures, we showed that FKBP65 does not undergo the unfolded protein response, a response associated with an upregulation of resident ER proteins that occurs after increased ER stress. When fibroblasts were treated with transforming growth factor (TGF)-beta1, which is upregulated during the development of pulmonary fibrosis and known to induce matrix production, FKBP65 expression and synthesis was also increased. Similar to type I collagen and tropoelastin, this response was completely inhibited in a dose-dependent manner by GGTI-298, a geranylgeranyl transferase I inhibitor. Treatment of fibroblasts with an inhibitor of ribonucleic acid (RNA) polymerase II after TGF-beta1 treatment showed that the effect of TGF-beta1 was not because of increased stabilization of the FKBP65 messenger RNA. In summary, we have shown that FKBP65 is highly expressed in lung development, downregulated in the adult, and can be reactivated in a coordinated manner with extracellular matrix proteins after lung injury. The expression pattern of FKBP65, which is atypical for general ER foldases, suggests that FKBP65 has a distinct set of developmentally regulated protein ligands. The response to injury, which may be in part a direct response to TGF-beta1, assures the presence of FKBP65 in the ER of cells actively producing components of the extracellular matrix.


Assuntos
Retículo Endoplasmático/enzimologia , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Imunofilinas/metabolismo , Pulmão/patologia , Peptidilprolil Isomerase/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose/patologia , Humanos , Imunofilinas/genética , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Peptidilprolil Isomerase/genética , Estabilidade de RNA , Proteínas de Ligação a Tacrolimo/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA