Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 35(4): 911-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25614286

RESUMO

OBJECTIVE: Studies of mice with mild Marfan syndrome (MFS) have correlated the development of thoracic aortic aneurysm (TAA) with improper stimulation of noncanonical (Erk-mediated) TGFß signaling by the angiotensin type I receptor (AT1r). This correlation was largely based on comparable TAA modifications by either systemic TGFß neutralization or AT1r antagonism. However, subsequent investigations have called into question some key aspects of this mechanism of arterial disease in MFS. To resolve these controversial points, here we made a head-to-head comparison of the therapeutic benefits of TGFß neutralization and AT1r antagonism in mice with progressively severe MFS (Fbn1(mgR/mgR) mice). APPROACH AND RESULTS: Aneurysm growth, media degeneration, aortic levels of phosphorylated Erk and Smad proteins and the average survival of Fbn1(mgR/mgR) mice were compared after a ≈3-month-long treatment with placebo and either the AT1r antagonist losartan or the TGFß-neutralizing antibody 1D11. In contrast to the beneficial effect of losartan, TGFß neutralization either exacerbated or mitigated TAA formation depending on whether treatment was initiated before (postnatal day 16; P16) or after (P45) aneurysm formation, respectively. Biochemical evidence-related aneurysm growth with Erk-mediated AT1r signaling, and medial degeneration with TGFß hyperactivity that was in part AT1r dependent. Importantly, P16-initiated treatment with losartan combined with P45-initiated administration of 1D11 prevented death of Fbn1(mgR/mgR) mice from ruptured TAA. CONCLUSIONS: By demonstrating that promiscuous AT1r and TGFß drive partially overlapping processes of arterial disease in MFS mice, our study argues for a therapeutic strategy against TAA that targets both signaling pathways although sparing the early protective role of TGFß.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Anticorpos Neutralizantes/farmacologia , Aorta Torácica/efeitos dos fármacos , Aneurisma da Aorta Torácica/prevenção & controle , Losartan/farmacologia , Síndrome de Marfan/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/patologia , Ruptura Aórtica/genética , Ruptura Aórtica/metabolismo , Ruptura Aórtica/patologia , Ruptura Aórtica/prevenção & controle , Modelos Animais de Doenças , Progressão da Doença , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas dos Microfilamentos/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação , Fosforilação , Receptor Tipo 1 de Angiotensina/metabolismo , Proteína Smad2/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
2.
Hum Mol Genet ; 19(24): 4790-8, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20871099

RESUMO

Reduced bone mineral density (osteopenia) is a poorly characterized manifestation of pediatric and adult patients afflicted with Marfan syndrome (MFS), a multisystem disorder caused by structural or quantitative defects in fibrillin-1 that perturb tissue integrity and TGFß bioavailability. Here we report that mice with progressively severe MFS (Fbn1(mgR/mgR) mice) develop osteopenia associated with normal osteoblast differentiation and bone formation. In vivo and ex vivo experiments, respectively, revealed that adult Fbn1(mgR/mgR) mice respond more strongly to locally induced osteolysis and that Fbn1(mgR/mgR) osteoblasts stimulate pre-osteoclast differentiation more than wild-type cells. Greater osteoclastogenic potential of mutant osteoblasts was largely attributed to Rankl up-regulation secondary to improper TGFß activation and signaling. Losartan treatment, which lowers TGFß signaling and restores aortic wall integrity in mice with mild MFS, did not mitigate bone loss in Fbn1(mgR/mgR) mice even though it ameliorated vascular disease. Conversely, alendronate treatment, which restricts osteoclast activity, improved bone quality but not aneurysm progression in Fbn1(mgR/mgR) mice. Taken together, our findings shed new light on the pathogenesis of osteopenia in MFS, in addition to arguing for a multifaceted treatment strategy in this congenital disorder of the connective tissue.


Assuntos
Alendronato/uso terapêutico , Aneurisma Aórtico/complicações , Aneurisma Aórtico/tratamento farmacológico , Doenças Ósseas Metabólicas/complicações , Doenças Ósseas Metabólicas/tratamento farmacológico , Losartan/uso terapêutico , Síndrome de Marfan/complicações , Alendronato/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/fisiopatologia , Aneurisma Aórtico/fisiopatologia , Doenças Ósseas Metabólicas/fisiopatologia , Proteínas Morfogenéticas Ósseas/metabolismo , Reabsorção Óssea/complicações , Reabsorção Óssea/fisiopatologia , Modelos Animais de Doenças , Fibrilina-1 , Fibrilinas , Losartan/farmacologia , Síndrome de Marfan/tratamento farmacológico , Síndrome de Marfan/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Coluna Vertebral/fisiopatologia , Tomografia Computadorizada por Raios X , Fator de Crescimento Transformador beta/metabolismo
3.
Cell Tissue Res ; 344(3): 511-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21538048

RESUMO

Fibrillin-1 and fibrillin-2 are structural components of the extracellular matrix which are also involved in modulating local TGFß and BMP bioavailability. Loss of fibrillin-1 or fibrillin-2 is associated with perturbed osteoblast maturation principally as the result of unbalanced TGFß and BMP signaling. Here, we demonstrated that stable expression of small hairpin RNAs against fibrillin-1(Fbn1) or fibrillin-2 (Fbn2) transcripts in the clonal osteoprogenitor cell line Kusa-A1 led to the same phenotypic and molecular manifestations as germline Fbn1- or Fbn2-null mutations in primary calvarial osteoblast cultures. Proof-of-concept experiments are also presented showing that Fbn1- or Fbn2-silenced Kusa-A1 cell lines are suitable models to identify candidate determinants of osteogenesis which are under the control of extracellular microfibrils. Specific findings included: the inference of a potential role for fibrillin-1-mediated cell-matrix interactions in regulating Kusa-A1 proliferation; the possibility of fibrillin-2 involvement in modulating the activity of transcription factor Runx2 by restricting microRNA expression and/or processing; and the suggestion that fibrillin-1 and fibrillin-2 influence Notch signaling indirectly by differentially regulating BMP signaling. Collectively, the data reiterated the notion that fibrillin-1 and fibrillin-2 exert opposite effects on osteoblast differentiation through the discrete modulation of a broad network of interacting signaling molecules.


Assuntos
Matriz Extracelular/metabolismo , Proteínas dos Microfilamentos/deficiência , Osteoblastos/citologia , Osteoblastos/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais , Transfecção
4.
Arterioscler Thromb Vasc Biol ; 29(12): 2083-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19850904

RESUMO

OBJECTIVE: Even though elastin and fibrillin-1 are the major structural components of elastic fibers, mutations in elastin and fibrillin-1 lead to narrowing of large arteries in supravalvular aortic stenosis and dilation of the ascending aorta in Marfan syndrome, respectively. A genetic approach was therefore used here to distinguish the differential contributions of elastin and fibrillin-1 to arterial development and compliance. METHODS AND RESULTS: Key parameters of cardiovascular function were compared among adult mice haploinsufficient for elastin (Eln(+/-)), fibrillin-1 (Fbn1(+/-)), or both proteins (dHet). Physiological and morphological comparisons correlate elastin haploinsufficiency with increased blood pressure and vessel length and tortuosity in dHet mice, and fibrillin-1 haploinsufficiency with increased aortic diameter in the same mutant animals. Mechanical tests confirm that elastin and fibrillin-1 impart elastic recoil and tensile strength to the aortic wall, respectively. Additional ex vivo analyses demonstrate additive and overlapping contributions of elastin and fibrillin-1 to the material properties of vascular tissues. Lastly, light and electron microscopy evidence implicates fibrillin-1 in the hypertension-promoted remodeling of the elastin-deficient aorta. CONCLUSIONS: These results demonstrate that elastin and fibrillin-1 have both differential and complementary roles in arterial wall formation and function, and advance our knowledge of the structural determinants of vascular physiology and disease.


Assuntos
Artérias/crescimento & desenvolvimento , Artérias/fisiologia , Tecido Elástico/fisiologia , Animais , Estenose Aórtica Supravalvular/etiologia , Estenose Aórtica Supravalvular/fisiopatologia , Artérias/patologia , Artérias/fisiopatologia , Fenômenos Biomecânicos/fisiologia , Complacência (Medida de Distensibilidade)/fisiologia , Modelos Animais de Doenças , Tecido Elástico/patologia , Tecido Elástico/fisiopatologia , Elastina/deficiência , Elastina/genética , Elastina/fisiologia , Matriz Extracelular/metabolismo , Fibrilina-1 , Fibrilinas , Humanos , Síndrome de Marfan/etiologia , Síndrome de Marfan/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Fenótipo
5.
Connect Tissue Res ; 49(1): 1-6, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18293172

RESUMO

Fibrillin-rich microfibrils have emerged recently as an informative model system in which to study fundamental questions related to extracellular matrix biology and connective tissue pathophysiology. As a result, these studies have yielded novel clinical concepts and promising therapeutic strategies. These achievements have been based on the realization from studies of genetically engineered mice that mutations in fibrillin-rich microfibrils impair both the structural integrity of connective tissues and signaling events by TGF-beta/BMP superfamily members. In this view, fibrillin-rich microfibrils represent architectural assemblies that specify the concentration and timely release of local effectors of morphogenesis and tissue remodeling, in addition to conferring structural integrity to individual organ systems. This review summarizes the evidence supporting our current understanding of the structural and instructive roles that fibrillin-rich microfibrils play during embryonic development and in human diseases.


Assuntos
Tecido Conjuntivo/fisiopatologia , Desenvolvimento Embrionário/fisiologia , Matriz Extracelular/fisiologia , Mamíferos/fisiologia , Microfibrilas/genética , Proteínas dos Microfilamentos/fisiologia , Animais , Aorta/citologia , Aorta/fisiologia , Osso e Ossos/embriologia , Osso e Ossos/fisiologia , Fibrilinas , Microfibrilas/fisiologia
6.
J Clin Invest ; 124(3): 1329-39, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531548

RESUMO

Patients with Marfan syndrome (MFS), a multisystem disorder caused by mutations in the gene encoding the extracellular matrix (ECM) protein fibrillin 1, are unusually vulnerable to stress-induced cardiac dysfunction. The prevailing view is that MFS-associated cardiac dysfunction is the result of aortic and/or valvular disease. Here, we determined that dilated cardiomyopathy (DCM) in fibrillin 1-deficient mice is a primary manifestation resulting from ECM-induced abnormal mechanosignaling by cardiomyocytes. MFS mice displayed spontaneous emergence of an enlarged and dysfunctional heart, altered physical properties of myocardial tissue, and biochemical evidence of chronic mechanical stress, including increased angiotensin II type I receptor (AT1R) signaling and abated focal adhesion kinase (FAK) activity. Partial fibrillin 1 gene inactivation in cardiomyocytes was sufficient to precipitate DCM in otherwise phenotypically normal mice. Consistent with abnormal mechanosignaling, normal cardiac size and function were restored in MFS mice treated with an AT1R antagonist and in MFS mice lacking AT1R or ß-arrestin 2, but not in MFS mice treated with an angiotensin-converting enzyme inhibitor or lacking angiotensinogen. Conversely, DCM associated with abnormal AT1R and FAK signaling was the sole abnormality in mice that were haploinsufficient for both fibrillin 1 and ß1 integrin. Collectively, these findings implicate fibrillin 1 in the physiological adaptation of cardiac muscle to elevated workload.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Síndrome de Marfan/metabolismo , Mecanotransdução Celular , Adulto , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Criança , Estudos Transversais , Matriz Extracelular/metabolismo , Fibrilina-1 , Fibrilinas , Quinase 1 de Adesão Focal/metabolismo , Humanos , Losartan/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Síndrome de Marfan/complicações , Síndrome de Marfan/patologia , Síndrome de Marfan/fisiopatologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Tamanho do Órgão , Receptor Tipo 1 de Angiotensina/metabolismo
7.
J Cell Biol ; 190(6): 1107-21, 2010 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-20855508

RESUMO

Extracellular regulation of signaling by transforming growth factor (TGF)-ß family members is emerging as a key aspect of organ formation and tissue remodeling. In this study, we demonstrate that fibrillin-1 and -2, the structural components of extracellular microfibrils, differentially regulate TGF-ß and bone morphogenetic protein (BMP) bioavailability in bone. Fibrillin-2-null (Fbn2(-/-)) mice display a low bone mass phenotype that is associated with reduced bone formation in vivo and impaired osteoblast maturation in vitro. This Fbn2(-/-) phenotype is accounted for by improper activation of latent TGF-ß that selectively blunts expression of osterix, the transcriptional regulator of osteoblast maturation, and collagen I, the structural template for bone mineralization. Cultured osteoblasts from Fbn1(-/-) mice exhibit improper latent TGF-ß activation as well, but mature faster because of increased availability of otherwise matrix-bound BMPs. Additional in vitro evidence excludes a direct role of microfibrils in supporting mineral deposition. Together, these findings identify the extracellular microfibrils as critical regulators of bone formation through the modulation of endogenous TGF-ß and BMP signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Osteogênese/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Disponibilidade Biológica , Matriz Óssea/metabolismo , Matriz Óssea/patologia , Calcificação Fisiológica/fisiologia , Diferenciação Celular , Células Cultivadas , Colágeno/metabolismo , Regulação para Baixo , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Humanos , Camundongos , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/deficiência , Modelos Biológicos , Tamanho do Órgão , Osteoblastos/metabolismo , Osteoblastos/patologia , Transdução de Sinais , Fator de Transcrição Sp7 , Fatores de Transcrição/metabolismo
8.
J Biol Chem ; 284(9): 5630-6, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19109253

RESUMO

Excessive transforming growth factor-beta (TGF-beta) signaling characterizes the progression of aortic aneurysm in mouse models of Marfan syndrome, a systemic disorder of the connective tissue that is caused by mutations in the gene encoding the extracellular matrix protein fibrillin-1. Fibrillin-1 mutations are believed to promote abnormal Smad2/3 signaling by impairing the sequestration of latent TGF-beta complexes into the extracellular matrix. Here we report that promiscuous Smad2/3 signaling is the cell-autonomous phenotype of primary cultures of vascular smooth muscle cells (VSMC) explanted from the thoracic aortas of Fbn1 mutant mice with either neonatal onset or progressively severe aortic aneurysm. This cellular phenotype was characterized in VSMC isolated from Fbn1-null (mgN/mgN) mice, which recapitulate the most severe form of Marfan syndrome. We found that loss of fibrillin-1 deposition promotes the production of intracellular reactive oxygen species and abnormal accumulation of phosphorylated TGF-beta-activated kinase 1 and p38 MAPK, in addition to increasing the levels of endogenous phospho-Smad2. We showed that improper Smad2/3 signaling in Fbn1-null VSMC is in part stimulated by phospho-p38 MAPK, which is in turn activated in response to signals other than those mediated by the kinase activity of the ALK5 receptor. Consistent with these cell culture data, in vivo analyses documented that phospho-p38 MAPK accumulates earlier than phospho-Smad2 in the aortic wall of mgN/mgN mice and that systemic inhibition of phospho-p38 MAPK activity lowers the levels of phospho-Smad2 in this tissue. Collectively, these findings indicate that improper activation of p38 MAPK is a precursor of constitutive Smad2/3 signaling in the aortic wall of a mouse model of neonatal lethal Marfan syndrome.


Assuntos
Aorta Torácica/metabolismo , Proteínas dos Microfilamentos/fisiologia , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Aorta Torácica/citologia , Células Cultivadas , Fibrilina-1 , Fibrilinas , Regulação da Expressão Gênica no Desenvolvimento , Immunoblotting , Técnicas Imunoenzimáticas , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2/genética , Proteína Smad3/genética
9.
J Biol Chem ; 281(12): 8016-23, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16407178

RESUMO

Fibrillin-rich microfibrils are extracellular assemblies that impart structural properties to the connective tissue. To elucidate the contribution of fibrillin-rich microfibrils to organogenesis, we have examined the vascular phenotype of a newly created strain of mice that completely lacks fibrillin-1 and the consequences of combined deficiency of fibrillins 1 and 2 on tissue formation. The results demonstrated that fibrillins 1 and 2 perform partially overlapping functions during aortic development. Fbn1-/- mice died soon after birth from ruptured aortic aneurysm, impaired pulmonary function, and/or diaphragmatic collapse. Analysis of the neonatal Fbn1-/- aorta documented a disorganized and poorly developed medial layer but normal levels of elastin cross-links. Transcriptional profiling revealed that aneurysm progression in Fbn1 null mice is accompanied by unproductive up-regulation of gene products normally involved in tissue repair and vascular integrity, such as plasminogen activator inhibitor-1, activin A, and cysteine-rich angiogenic protein 61. In contrast to Fbn1-/- mice, Fbn2 null mice had a well developed and morphologically normal aortic wall. However, virtually all Fbn1-/-;Fbn2-/- embryos and about half of the Fbn1+/-;Fbn2-/- embryos died in utero and displayed a significantly more severe vascular phenotype than Fbn1-/- mice. Consistent with a specialized function of fibrillin-2, electron microscopy visualized ultrastructurally different microfibrils in Fbn1 null compared with control cell cultures. Collectively, these data demonstrate that involvement of fibrillin-2 in the initial assembly of the aortic matrix overlaps in part with fibrillin-1 and that continued fibrillin-1 deposition is absolutely required for the maturation and function of the vessel during neonatal life.


Assuntos
Aorta/embriologia , Proteínas dos Microfilamentos/química , Ativinas/metabolismo , Alelos , Animais , Células Cultivadas , Proteínas Contráteis/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , Cruzamentos Genéticos , Proteína Rica em Cisteína 61 , Desmosina/metabolismo , Elastina/metabolismo , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Fibroblastos/metabolismo , Genótipo , Heterozigoto , Proteínas Imediatamente Precoces/metabolismo , Immunoblotting , Subunidades beta de Inibinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Transgênicos , Microfibrilas/química , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica , Modelos Genéticos , Fenótipo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , RNA/metabolismo
10.
Science ; 312(5770): 117-21, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16601194

RESUMO

Aortic aneurysm and dissection are manifestations of Marfan syndrome (MFS), a disorder caused by mutations in the gene that encodes fibrillin-1. Selected manifestations of MFS reflect excessive signaling by the transforming growth factor-beta (TGF-beta) family of cytokines. We show that aortic aneurysm in a mouse model of MFS is associated with increased TGF-beta signaling and can be prevented by TGF-beta antagonists such as TGF-beta-neutralizing antibody or the angiotensin II type 1 receptor (AT1) blocker, losartan. AT1 antagonism also partially reversed noncardiovascular manifestations of MFS, including impaired alveolar septation. These data suggest that losartan, a drug already in clinical use for hypertension, merits investigation as a therapeutic strategy for patients with MFS and has the potential to prevent the major life-threatening manifestation of this disorder.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Aneurisma Aórtico/prevenção & controle , Modelos Animais de Doenças , Losartan/uso terapêutico , Síndrome de Marfan/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo , Antagonistas Adrenérgicos beta/administração & dosagem , Antagonistas Adrenérgicos beta/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Anticorpos/imunologia , Aorta/patologia , Aneurisma Aórtico/etiologia , Tecido Elástico/patologia , Feminino , Fibrilina-1 , Fibrilinas , Losartan/administração & dosagem , Pulmão/patologia , Pneumopatias/tratamento farmacológico , Pneumopatias/patologia , Síndrome de Marfan/complicações , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patologia , Camundongos , Proteínas dos Microfilamentos/genética , Mutação , Testes de Neutralização , Gravidez , Complicações na Gravidez/tratamento farmacológico , Propranolol/administração & dosagem , Propranolol/uso terapêutico , Alvéolos Pulmonares/patologia , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/imunologia
11.
Birth Defects Res A Clin Mol Teratol ; 73(3): 133-5, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15751030

RESUMO

The synthetic estrogen diethylstilbestrol (DES) was administered to pregnant women between the 1940s and the mid-1970s and is believed to be responsible for numerous uterine/cervical/vaginal malformations and cancers that appeared after birth and in young adult life. This medical tragedy has served as one of the prototypical examples of a phenomenon known as "endocrine disruption," in which either environmental agents or other compounds disrupt normal hormonal signaling in the body. Whereas DES signals through estrogen receptors, the subsequent molecular targets were largely unknown. We had identified Wnt7a as a target in this pathway and have used genetic analyses of mutant mice to demonstrate that disruption of Wnt7a is the key event leading to the DES phenotypes and cancers. We find that Wnt7a expression is only transiently deregulated in response to DES exposure, leading to the conclusion that critical events during early reproductive tract development results in a permanent change or "reprogramming" in subsequent development.


Assuntos
Dietilestilbestrol/efeitos adversos , Estrogênios não Esteroides/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal , Proteínas Proto-Oncogênicas/metabolismo , Animais , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Camundongos , Neoplasias/congênito , Neoplasias/etiologia , Gravidez , Proteínas Wnt
12.
Am J Med Genet C Semin Med Genet ; 139C(1): 4-9, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16273535

RESUMO

Marfan syndrome (MFS) is a systemic disorder of the connective tissue that is inherited as an autosomal dominant trait and which displays variable manifestations in the ocular, skeletal, and cardiovascular systems. These pleiotropic manifestations are accounted for by mutations in fibrillin-1, the building block of extracellular microfibrils. During the past 10 years, we have witnessed significant progress in delineating the pathological events responsible for the manifestations of MFS. Much of this progress has been based on the creation and analysis of fibrillin-1 mutant mouse lines that faithfully recapitulate the spectrum of clinical severity of MFS. These studies have established the critical contribution of fibrillin-1 deficiency to disease progression through altered cell-matrix interactions and dysregulated TGF-beta signaling. As a result, our definition of MFS as the prototypical structural disorder of the connective tissue has changed to that of a developmental abnormality with broad and complex effects on the morphogenesis and function of multiple organ systems. Importantly, new biological targets have emerged that may yield exciting new opportunities for the development of productive treatment strategies in MFS.


Assuntos
Síndrome de Marfan/genética , Síndrome de Marfan/patologia , Microfibrilas/patologia , Proteínas dos Microfilamentos/genética , Fenótipo , Transdução de Sinais/genética , Animais , Fibrilina-1 , Fibrilinas , Humanos , Camundongos , Mutação/genética , Fator de Crescimento Transformador beta/metabolismo
13.
Biol Reprod ; 71(2): 444-54, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15070830

RESUMO

The murine female reproductive tract is undifferentiated at birth and undergoes pronounced growth and cytodifferentiation during postnatal life. Postnatal reproductive tract development proceeds in the absence of high levels of circulating estrogens and is disrupted by precocious exposure to estrogens. The WNT gene family is critical in guiding the epithelial-mesenchymal interactions that direct postnatal uterine development. We have previously described a role for Wnt7a in controlling morphogenesis in the uterus. In addition to patterning defects, Wnt7a mutant uteri are atrophic in adults and do not show robust postnatal growth. In the present study, we examine immature female Wnt7a mutant and wild-type uteri to assess the cellular processes that underlie this failure in postnatal uterine growth. Levels of proliferation are higher in wild-type versus Wnt7a mutant uteri. Exposure to the potent estrogen-agonist diethylstilbestrol (DES) leads to an increase in cell proliferation in the uterus in wild-type as well as in mutant uteri, indicating that Wnt7a is not required in mediating cell proliferation. In contrast, we observe that Wnt7a mutant uteri display high levels of cell death in response to DES, whereas wild-type uteri display almost no cell death, revealing that Wnt7a plays a key role as a cell death suppressor. The expression pattern of other key regulatory genes that guide uterine development, including estrogen receptor (alpha), Hox, and other WNT genes, reveals either abnormal spatial distribution of transcripts or abnormal regulation in response to DES exposure. Taken together, the results of the present study demonstrate that Wnt7a coordinates a variety of cell and developmental pathways that guide postnatal uterine growth and hormonal responses and that disruption of these pathways leads to aberrant cell death.


Assuntos
Morte Celular/fisiologia , Estrogênios/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Útero/crescimento & desenvolvimento , Útero/fisiologia , Animais , Divisão Celular , Dietilestilbestrol/farmacologia , Estrogênios não Esteroides/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Ductos Paramesonéfricos/crescimento & desenvolvimento , Ductos Paramesonéfricos/fisiologia , Proteínas Proto-Oncogênicas/genética , Útero/patologia , Proteínas Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA