Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Hum Mol Genet ; 31(19): 3281-3289, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-35567544

RESUMO

A disproportionate tall stature is the most evident manifestation in Marfan syndrome (MFS), a multisystem condition caused by mutations in the extracellular protein and TGFß modulator, fibrillin-1. Unlike cardiovascular manifestations, there has been little effort devoted to unravel the molecular mechanism responsible for long bone overgrowth in MFS. By combining the Cre-LoxP recombination system with metatarsal bone cultures, here we identify the outer layer of the perichondrium as the tissue responsible for long bone overgrowth in MFS mice. Analyses of differentially expressed genes in the fibrillin-1-deficient perichondrium predicted that loss of TGFß signaling may influence chondrogenesis in the neighboring epiphyseal growth plate (GP). Immunohistochemistry revealed that fibrillin-1 deficiency in the outer perichondrium is associated with decreased accumulation of latent TGFß-binding proteins (LTBPs)-3 and -4, and reduced levels of phosphorylated (activated) Smad2. Consistent with these findings, mutant metatarsal bones grown in vitro were longer and released less TGFß than the wild-type counterparts. Moreover, addition of recombinant TGFß1 normalized linear growth of mutant metatarsal bones. We conclude that longitudinal bone overgrowth in MFS is accounted for by diminished sequestration of LTBP-3 and LTBP-4 into the fibrillin-1-deficient matrix of the outer perichondrium, which results in less TGFß signaling locally and improper GP differentiation distally.


Assuntos
Síndrome de Marfan , Animais , Fibrilina-1/genética , Fibrilina-2 , Fibrilinas , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas de Ligação a TGF-beta Latente/metabolismo , Síndrome de Marfan/genética , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
2.
Am J Hum Genet ; 102(4): 706-712, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625025

RESUMO

The major diseases affecting the thoracic aorta are aneurysms and acute dissections, and pathogenic variants in 11 genes are confirmed to lead to heritable thoracic aortic disease. However, many families in which multiple members have thoracic aortic disease do not have alterations in the known aortopathy genes. Genes highly expressed in the aorta were assessed for rare variants in exome sequencing data from such families, and compound rare heterozygous variants (p.Pro45Argfs∗25 and p.Glu750∗) in LTBP3 were identified in affected members of one family. A homozygous variant (p.Asn678_Gly681delinsThrCys) that introduces an additional cysteine into an epidermal growth factor (EGF)-like domain in the corresponding protein, latent TGF-ß binding protein (LTBP-3), was identified in a second family. Individuals with compound heterozygous or homozygous variants in these families have aneurysms and dissections of the thoracic aorta, as well as aneurysms of the abdominal aorta and other arteries, along with dental abnormalities and short stature. Heterozygous carriers of the p.Asn678_Gly681delinsThrCys variant have later onset of thoracic aortic disease, as well as dental abnormalities. In these families, LTBP3 variants segregated with thoracic aortic disease with a combined LOD score of 3.9. Additionally, heterozygous rare LTBP3 variants were found in individuals with early onset of acute aortic dissections, and some of these variants disrupted LTBP-3 levels or EGF-like domains. When compared to wild-type mice, Ltbp3-/- mice have enlarged aortic roots and ascending aortas. In summary, homozygous LTBP3 pathogenic variants predispose individuals to thoracic aortic aneurysms and dissections, along with the previously described skeletal and dental abnormalities.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Predisposição Genética para Doença , Proteínas de Ligação a TGF-beta Latente/genética , Mutação/genética , Adulto , Idoso de 80 Anos ou mais , Animais , Pressão Sanguínea/genética , Feminino , Homozigoto , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Linhagem
3.
Proc Natl Acad Sci U S A ; 112(45): 14012-7, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26494287

RESUMO

Marfan syndrome (MFS) is an autosomal dominant disorder of connective tissue, caused by mutations of the microfibrillar protein fibrillin-1, that predisposes affected individuals to aortic aneurysm and rupture and is associated with increased TGFß signaling. TGFß is secreted from cells as a latent complex consisting of TGFß, the TGFß propeptide, and a molecule of latent TGFß binding protein (LTBP). Improper extracellular localization of the latent complex can alter active TGFß levels, and has been hypothesized as an explanation for enhanced TGFß signaling observed in MFS. We previously reported the absence of LTBP-3 in matrices lacking fibrillin-1, suggesting that perturbed TGFß signaling in MFS might be due to defective interaction of latent TGFß complexes containing LTBP-3 with mutant fibrillin-1 microfibrils. To test this hypothesis, we genetically suppressed Ltbp3 expression in a mouse model of progressively severe MFS. Here, we present evidence that MFS mice lacking LTBP-3 have improved survival, essentially no aneurysms, reduced disruption and fragmentation of medial elastic fibers, and decreased Smad2/3 and Erk1/2 activation in their aortas. These data suggest that, in MFS, improper localization of latent TGFß complexes composed of LTBP-3 and TGFß contributes to aortic disease progression.


Assuntos
Aneurisma da Aorta Torácica/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Síndrome de Marfan/complicações , Síndrome de Marfan/genética , Complexos Multiproteicos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Análise de Variância , Animais , Aneurisma da Aorta Torácica/etiologia , DNA Complementar/biossíntese , Fibrilina-1 , Fibrilinas , Imuno-Histoquímica , Proteínas de Ligação a TGF-beta Latente/deficiência , Camundongos , Proteínas dos Microfilamentos/genética , Músculo Liso Vascular/citologia , Reação em Cadeia da Polimerase em Tempo Real
4.
Hum Mol Genet ; 24(11): 3038-49, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25669657

RESUMO

Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Here, we report on four families, three of them consanguineous, with an identical phenotype, characterized by significant short stature with brachyolmia and hypoplastic amelogenesis imperfecta (AI) with almost absent enamel. This phenotype was first described in 1996 by Verloes et al. as an autosomal recessive form of brachyolmia associated with AI. Whole-exome sequencing resulted in the identification of recessive hypomorphic mutations including deletion, nonsense and splice mutations, in the LTBP3 gene, which is involved in the TGF-beta signaling pathway. We further investigated gene expression during mouse development and tooth formation. Differentiated ameloblasts synthesizing enamel matrix proteins and odontoblasts expressed the gene. Study of an available knockout mouse model showed that the mutant mice displayed very thin to absent enamel in both incisors and molars, hereby recapitulating the AI phenotype in the human disorder.


Assuntos
Amelogênese Imperfeita/genética , Proteínas de Ligação a TGF-beta Latente/genética , Osteocondrodisplasias/genética , Adolescente , Amelogênese Imperfeita/diagnóstico por imagem , Animais , Sequência de Bases , Criança , Consanguinidade , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Estudos de Associação Genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação de Sentido Incorreto , Osteocondrodisplasias/diagnóstico por imagem , Linhagem , Radiografia , Deleção de Sequência
5.
Biochem Biophys Res Commun ; 482(4): 1387-1392, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27956181

RESUMO

The four latent transforming growth factor-ß (TGF-ß) binding proteins LTBP1-4 are extracellular matrix-associated proteins playing a critical role in the activation of TGF-ß. The LTBP1 gene forms two major transcript variants (i.e. Ltbp1S and Ltbp1L) that are derived from different promoters. We have previously shown the importance of LTBP1 in vivo by using three different Ltbp1 null mice that were either deleted for exons 1 and 2 (Ltbp1L knockout), exon 5 (Ltbp1ΔEx5), or exon 8 (Ltbp1ΔEx8). While the Ltbp1L knockout and the Ltbp1ΔEx8 are perinatal lethal and die of cardiovascular abnormalities, the Ltbp1ΔEx5 is viable because it expresses a short form of Ltbp1L that lacks 55 amino acids (Δ55 variant of Ltbp1) formed by splicing out exon 5, while lacking the Ltbp1S variant. Since only the Ltbp1ΔEx5 mouse is viable, we have used this model to address aspects of puberty, fertility, age-dependent reproduction, and ovary function. We report for the first time a function of LTBP1 in female reproduction. The Ltbp1ΔEx5 females showed impaired fertility associated with delayed sexual maturity (p = 0.0074) and ovarian cyst formation in females older than 40 weeks (p = 0.0204).


Assuntos
Infertilidade Feminina/genética , Proteínas de Ligação a TGF-beta Latente/genética , Cistos Ovarianos/metabolismo , Processamento Alternativo , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Estrogênios/sangue , Éxons , Matriz Extracelular/metabolismo , Feminino , Fertilidade , Fertilização in vitro , Genótipo , Proteínas de Ligação a TGF-beta Latente/deficiência , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Progesterona/sangue
6.
Eur J Oral Sci ; 125(1): 8-17, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28084688

RESUMO

Latent-transforming growth factor beta-binding protein 3 (LTBP-3) is important for craniofacial morphogenesis and hard tissue mineralization, as it is essential for activation of transforming growth factor-ß (TGF-ß). To investigate the role of LTBP-3 in tooth formation we performed micro-computed tomography (micro-CT), histology, and scanning electron microscopy analyses of adult Ltbp3-/- mice. The Ltbp3-/- mutants presented with unique craniofacial malformations and reductions in enamel formation that began at the matrix formation stage. Organization of maturation-stage ameloblasts was severely disrupted. The lateral side of the incisor was affected most. Reduced enamel mineralization, modification of the enamel prism pattern, and enamel nodules were observed throughout the incisors, as revealed by scanning electron microscopy. Molar roots had internal irregular bulbous-like formations. The cementum thickness was reduced, and microscopic dentinal tubules showed minor nanostructural changes. Thus, LTBP-3 is required for ameloblast differentiation and for the formation of decussating enamel prisms, to prevent enamel nodule formation, and for proper root morphogenesis. Also, and consistent with the role of TGF-ß signaling during mineralization, almost all craniofacial bone components were affected in Ltbp3-/- mice, especially those involving the upper jaw and snout. This mouse model demonstrates phenotypic overlap with Verloes Bourguignon syndrome, also caused by mutation of LTBP3, which is hallmarked by craniofacial anomalies and amelogenesis imperfecta phenotypes.


Assuntos
Amelogênese/genética , Esmalte Dentário/anormalidades , Proteínas de Ligação a TGF-beta Latente/genética , Ameloblastos/metabolismo , Amelogênese Imperfeita/genética , Animais , Esmalte Dentário/ultraestrutura , Genótipo , Masculino , Camundongos , Camundongos Mutantes , Microscopia Eletrônica de Varredura , Mutação , Osteocondrodisplasias/genética , Fenótipo , Calcificação de Dente/genética , Fator de Crescimento Transformador beta/genética , Microtomografia por Raio-X
7.
Proc Natl Acad Sci U S A ; 110(8): 2852-7, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382201

RESUMO

Elastic fiber assembly requires deposition of elastin monomers onto microfibrils, the mechanism of which is incompletely understood. Here we show that latent TGF-ß binding protein 4 (LTBP-4) potentiates formation of elastic fibers through interacting with fibulin-5, a tropoelastin-binding protein necessary for elastogenesis. Decreased expression of LTBP-4 in human dermal fibroblast cells by siRNA treatment abolished the linear deposition of fibulin-5 and tropoelastin on microfibrils. It is notable that the addition of recombinant LTBP-4 to cell culture medium promoted elastin deposition on microfibrils without changing the expression of elastic fiber components. This elastogenic property of LTBP-4 is independent of bound TGF-ß because TGF-ß-free recombinant LTBP-4 was as potent an elastogenic inducer as TGF-ß-bound recombinant LTBP-4. Without LTBP-4, fibulin-5 and tropoelastin deposition was discontinuous and punctate in vitro and in vivo. These data suggest a unique function for LTBP-4 during elastic fibrogenesis, making it a potential therapeutic target for elastic fiber regeneration.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Proteínas de Ligação a TGF-beta Latente/fisiologia , Proteínas Recombinantes/metabolismo , Animais , Células HEK293 , Humanos , Proteínas de Ligação a TGF-beta Latente/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , Interferência de RNA
8.
J Cell Physiol ; 230(1): 226-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24962333

RESUMO

Mice deficient in Latent TGFß Binding Protein 4 (Ltbp4) display a defect in lung septation and elastogenesis. The lung septation defect is normalized by genetically decreasing TGFß2 levels. However, the elastic fiber assembly is not improved in Tgfb2(-/-) ;Ltbp4S(-/-) compared to Ltbp4S(-/-) lungs. We found that decreased levels of TGFß1 or TGFß3 did not improve lung septation indicating that the TGFß isoform elevated in Ltbp4S(-/-) lungs is TGFß2. Expression of a form of Ltbp4 that could not bind latent TGFß did not affect lung phenotype indicating that normal lung development does not require the formation of LTBP4-latent TGFß complexes. Therefore, the change in TGFß-level in the lungs is not directly related to Ltbp4 deficiency but probably is a consequence of changes in the extracellular matrix. Interestingly, combination of the Ltbp4S(-/-) mutation with a fibulin-5 null mutant in Fbln5(-/-) ;Ltbp4S(-/-) mice improves the lung septation compared to Ltbp4S(-/-) lungs. Large globular elastin aggregates characteristic for Ltbp4S(-/-) lungs do not form in Fbln5(-/-) ;Ltbp4S(-/-) lungs and EM studies showed that elastic fibers in Fbln5(-/-) ;Ltbp4S(-/-) lungs resemble those found in Fbln5(-/-) mice. These results are consistent with a role for TGFß2 in lung septation and for Ltbp4 in regulating fibulin-5 dependent elastic fiber assembly.


Assuntos
Padronização Corporal/genética , Tecido Elástico/embriologia , Proteínas da Matriz Extracelular/fisiologia , Proteínas de Ligação a TGF-beta Latente/fisiologia , Pulmão/embriologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Tecido Elástico/anormalidades , Elastina/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibrilinas , Proteínas de Ligação a TGF-beta Latente/genética , Pulmão/anormalidades , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Proteínas Recombinantes/genética , Transdução de Sinais/genética , Fator de Crescimento Transformador beta2/genética
9.
Nat Genet ; 37(3): 275-81, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731757

RESUMO

We report heterozygous mutations in the genes encoding either type I or type II transforming growth factor beta receptor in ten families with a newly described human phenotype that includes widespread perturbations in cardiovascular, craniofacial, neurocognitive and skeletal development. Despite evidence that receptors derived from selected mutated alleles cannot support TGFbeta signal propagation, cells derived from individuals heterozygous with respect to these mutations did not show altered kinetics of the acute phase response to administered ligand. Furthermore, tissues derived from affected individuals showed increased expression of both collagen and connective tissue growth factor, as well as nuclear enrichment of phosphorylated Smad2, indicative of increased TGFbeta signaling. These data definitively implicate perturbation of TGFbeta signaling in many common human phenotypes, including craniosynostosis, cleft palate, arterial aneurysms, congenital heart disease and mental retardation, and suggest that comprehensive mechanistic insight will require consideration of both primary and compensatory events.


Assuntos
Receptores de Ativinas Tipo I/genética , Desenvolvimento Ósseo/genética , Sistema Cardiovascular/crescimento & desenvolvimento , Transtornos Cognitivos/genética , Face , Mutação , Receptores de Fatores de Crescimento Transformadores beta/genética , Crânio/crescimento & desenvolvimento , Sequência de Aminoácidos , Pré-Escolar , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Fenótipo , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Homologia de Sequência de Aminoácidos , Síndrome
10.
Matrix Biol Plus ; 22: 100149, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38831847

RESUMO

Although the mechanism for activation of latent TGFß1 and TGFß3 is understood to involve the binding of the TGFß propeptide (LAP) to both an integrin and an insoluble substrate, the activation of latent TGFß2 has been unclear because the TGFß2 LAP does not have the classical integrin binding sequence found in the other two TGFß isoform LAPs. To assess the potential requirement for covalent linkage with a matrix or cell surface protein for the activation of latent TGFß2, we generated mice in which the TGFß2 Cys residue predicted to be involved in binding was mutated to Ser (Tgfb2C24S). We reasoned that, if covalent interaction with a second molecule is required for latent TGFß2 activation, mutant mice should display a Tgfb2 null (Tgfb2-/-)-like phenotype. Tgfb2C24S mice closely phenocopy Tgfb2-/- mice with death in utero between E18 and P1 and with congenital heart and kidney defects similar to those described for Tgfb2-/- mice. The mutant latent TGFß2 is secreted at levels similar to WT, yet TGFß signaling monitored as nuclear pSmad2 is suppressed. We conclude that, like latent TGFß1, latent TGFß2 activation requires binding to an immobilized matrix or plasma membrane molecule.

11.
JACC Basic Transl Sci ; 9(2): 185-199, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38510715

RESUMO

The severity of aortic stenosis (AS) is associated with acquired von Willebrand syndrome (AVWS) and gastrointestinal bleeding, leading to anemia (Heyde's syndrome). We investigated how anemia is linked with AS and AVWS using the LA100 mouse model and patients with AS. Induction of anemia in LA100 mice increased transforming growth factor (TGF)-ß1 activation, AVWS, and AS progression. Patients age >75 years with severe AS had higher plasma TGF-ß1 levels and more severe anemia than AS patients age <75 years, and there was a correlation between TGF-ß1 and anemia. These data are compatible with the hypothesis that the blood loss anemia of Heyde's syndrome contributes to AS progression via WSS-induced activation of platelet TGF-ß1 and additional gastrointestinal bleeding via WSS-induced AVWS.

12.
Matrix Biol ; 121: 41-55, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37217119

RESUMO

To assess the contribution of individual TGF-ß isoforms to aortopathy in Marfan syndrome (MFS), we quantified the survival and phenotypes of mice with a combined fibrillin1 (the gene defective in MFS) hypomorphic mutation and a TGF-ß1, 2, or 3 heterozygous null mutation. The loss of TGF-ß2, and only TGF-ß2, resulted in 80% of the double mutant animals dying earlier, by postnatal day 20, than MFS only mice. Death was not from thoracic aortic rupture, as observed in MFS mice, but was associated with hyperplastic aortic valve leaflets, aortic regurgitation, enlarged aortic root, increased heart weight, and impaired lung alveolar septation. Thus, there appears to be a relationship between loss of fibrillin1 and TGF-ß2 in the postnatal development of the heart, aorta and lungs.


Assuntos
Haploinsuficiência , Síndrome de Marfan , Animais , Camundongos , Aorta , Fibrilina-1/genética , Síndrome de Marfan/genética , Fenótipo , Fator de Crescimento Transformador beta2/genética
13.
bioRxiv ; 2023 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38586035

RESUMO

Collagen VI-related disorders (COL6-RDs) are a group of rare muscular dystrophies caused by pathogenic variants in collagen VI genes (COL6A1, COL6A2, and COL6A3). Collagen type VI is a heterotrimeric, microfibrillar component of the muscle extracellular matrix (ECM), predominantly secreted by resident fibroadipogenic precursor cells in skeletal muscle. The absence or mislocalizatoion of collagen VI in the ECM underlies the non-cell autonomous dysfunction and dystrophic changes in skeletal muscle with an as of yet elusive direct mechanistic link between the ECM and myofiber dysfunction. Here, we conduct a comprehensive natural history and outcome study in a novel mouse model of COL6-RDs (Col6a2-/- mice) using standardized (Treat-NMD) functional, histological, and physiologic parameter. Notably, we identify a conspicuous dysregulation of the TGFß pathway early in the disease process and propose that the collagen VI deficient matrix is not capable of regulating the dynamic TGFß bioavailability at baseline and also in response to muscle injury. Thus, we propose a new mechanism for pathogenesis of the disease that links the ECM regulation of TGFß with downstream skeletal muscle abnormalities, paving the way for developing and validating therapeutics that target this pathway.

14.
J Cell Physiol ; 227(12): 3828-36, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22495824

RESUMO

Fibrillin microfibrils are extracellular matrix structures with essential functions in the development and the organization of tissues including blood vessels, bone, limbs and the eye. Fibrillin-1 and fibrillin-2 form the core of fibrillin microfibrils, to which multiple proteins associate to form a highly organized structure. Defining the components of this structure and their interactions is crucial to understand the pathobiology of microfibrillopathies associated with mutations in fibrillins and in microfibril-associated molecules. In this study, we have analyzed both in vitro and in vivo the role of fibrillin microfibrils in the matrix deposition of latent TGF-ß binding protein 1 (LTBP-1), -3 and -4; the three LTBPs that form a complex with TGF-ß. In Fbn1(-/-) ascending aortas and lungs, LTBP-3 and LTBP-4 are not incorporated into a matrix lacking fibrillin-1 microfibrils, whereas LTBP-1 is still deposited. In addition, in cultures of Fbn1(-/-) smooth muscle cells or lung fibroblasts, LTBP-3 and LTBP-4 are not incorporated into a matrix lacking fibrillin-1 microfibrils, whereas LTBP-1 is still deposited. Fibrillin-2 is not involved in the deposition of LTBP-1 in Fbn1(-/-) extracellular matrix as cells deficient for both fibrillin-1 and fibrillin-2 still incorporate LTBP-1 in their matrix. However, blocking the formation of the fibronectin network in Fbn1(-/-) cells abrogates the deposition of LTBP-1. Together, these data indicate that LTBP-3 and LTBP-4 association with the matrix depends on fibrillin-1 microfibrils, whereas LTBP-1 association depends on a fibronectin network.


Assuntos
Fibronectinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Ligação a TGF-beta Latente/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , DNA Complementar/genética , DNA Complementar/metabolismo , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Fibroblastos/metabolismo , Fibronectinas/genética , Proteínas de Ligação a TGF-beta Latente/genética , Pulmão/citologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , RNA/genética , RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
15.
J Cell Biochem ; 113(2): 410-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22223425

RESUMO

Latent transforming growth factor beta (TGF-ß) binding proteins (LTBPs) are large extracellular glycoproteins structurally similar to fibrillins. They perform intricate and important roles in the extracellular matrix (ECM) and perturbations of their function manifest as a wide range of diseases. LTBPs are major regulators of TGF-ß bioavailability and action. In addition, LTBPs interact with other ECM proteins-from cytokines to large multi-factorial aggregates like microfibrils and elastic fibers, affecting their genesis, structure, and performance. In the present article, we review recent advancements in the field and relate the complex roles of LTBP in development and homeostasis.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Animais , Matriz Extracelular/metabolismo , Doenças Genéticas Inatas/genética , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Mutação , Ligação Proteica , Estrutura Terciária de Proteína
16.
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
17.
Am J Hum Genet ; 85(5): 593-605, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19836010

RESUMO

We report recessive mutations in the gene for the latent transforming growth factor-beta binding protein 4 (LTBP4) in four unrelated patients with a human syndrome disrupting pulmonary, gastrointestinal, urinary, musculoskeletal, craniofacial, and dermal development. All patients had severe respiratory distress, with cystic and atelectatic changes in the lungs complicated by tracheomalacia and diaphragmatic hernia. Three of the four patients died of respiratory failure. Cardiovascular lesions were mild, limited to pulmonary artery stenosis and patent foramen ovale. Gastrointestinal malformations included diverticulosis, enlargement, tortuosity, and stenosis at various levels of the intestinal tract. The urinary tract was affected by diverticulosis and hydronephrosis. Joint laxity and low muscle tone contributed to musculoskeletal problems compounded by postnatal growth delay. Craniofacial features included microretrognathia, flat midface, receding forehead, and wide fontanelles. All patients had cutis laxa. Four of the five identified LTBP4 mutations led to premature termination of translation and destabilization of the LTBP4 mRNA. Impaired synthesis and lack of deposition of LTBP4 into the extracellular matrix (ECM) caused increased transforming growth factor-beta (TGF-beta) activity in cultured fibroblasts and defective elastic fiber assembly in all tissues affected by the disease. These molecular defects were associated with blocked alveolarization and airway collapse in the lung. Our results show that coupling of TGF-beta signaling and ECM assembly is essential for proper development and is achieved in multiple human organ systems by multifunctional proteins such as LTBP4.


Assuntos
Derme/anormalidades , Intestinos/anormalidades , Proteínas de Ligação a TGF-beta Latente/genética , Pulmão/anormalidades , Mutação , Sistema Urinário/anormalidades , Células Cultivadas , Criança , Pré-Escolar , Técnicas de Cocultura , Meios de Cultivo Condicionados/química , DNA/genética , DNA/isolamento & purificação , Derme/metabolismo , Derme/ultraestrutura , Feminino , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Homozigoto , Humanos , Imuno-Histoquímica , Lactente , Mucosa Intestinal/metabolismo , Proteínas de Ligação a TGF-beta Latente/química , Pulmão/metabolismo , Masculino , Sistema Musculoesquelético , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Pele/citologia , Síndrome , Sistema Urinário/metabolismo
18.
Dev Dyn ; 240(1): 176-87, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21181942

RESUMO

Transforming Growth Factor ß (TGF-ß) is crucial for valve development and homeostasis. The long form of Latent TGF-ß binding protein 1 (LTBP1L) covalently binds all TGF-ß isoforms and regulates their bioavailability. Ltbp1L expression analysis during valvulogenesis revealed two patterns of Ltbp1L production: an early one (E9.5-11.5) associated with endothelial-to-mesenchymal transformation (EMT); and a late one (E12.5 to birth) contemporaneous with valve remodeling. Similarly, histological analysis of Ltbp1L(-/-) developing valves identified two different pathologies: generation of hypoplastic endocardial cushions in early valvulogenesis, followed by development of hyperplastic valves in late valvulogenesis. Ltbp1L promotes valve EMT, as Ltbp1L absence yields hypoplastic endocardial cushions in vivo and attenuated EMT in vitro. Ltbp1L(-/-) valve hyperplasia in late valvuogenesis represents a consequence of prolonged EMT. We demonstrate that Ltbp1L is a major regulator of Tgf-ß activity during valvulogenesis since its absence results in a perturbed Tgf-ß pathway that causes all Ltbp1L(-/-) valvular defects.


Assuntos
Valvas Cardíacas/embriologia , Proteínas de Ligação a TGF-beta Latente/fisiologia , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Valvas Cardíacas/anormalidades , Valvas Cardíacas/metabolismo , Hiperplasia , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Valva Mitral/anormalidades , Valva Mitral/embriologia , Valva Mitral/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Crescimento Transformadores/genética , Fatores de Crescimento Transformadores/metabolismo
19.
J Biol Chem ; 285(26): 20242-51, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20404337

RESUMO

Fibrillin microfibrils are polymeric structures present in connective tissues. The importance of fibrillin microfibrils to connective tissue function has been demonstrated by the multiple genetic disorders caused by mutations in fibrillins and in microfibril-associated molecules. However, knowledge of microfibril structure is limited, largely due to their insolubility. Most previous studies have focused on how fibrillin-1 is organized within microfibril polymers. In this study, an immunochemical approach was used to circumvent the insolubility of microfibrils to determine the role of fibrillin-2 in postnatal microfibril structure. Results obtained from studies of wild type and fibrillin-1 null tissues, using monoclonal and polyclonal antibodies with defined epitopes, demonstrated that N-terminal fibrillin-2 epitopes are masked in postnatal microfibrils and can be revealed by enzymatic digestion or by genetic ablation of Fbn1. From these studies, we conclude that fetal fibrillin polymers form an inner core within postnatal microfibrils and that microfibril structure evolves as growth and development proceed into the postnatal period. Furthermore, documentation of a novel cryptic site present in EGF4 in fibrillin-1 underscores the molecular complexity and tissue-specific differences in microfibril structure.


Assuntos
Microfibrilas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sequência de Aminoácidos , Âmnio/química , Animais , Anticorpos Monoclonais/imunologia , Embrião de Galinha , Ectoderma/embriologia , Ectoderma/metabolismo , Epitopos/imunologia , Extremidades/embriologia , Feminino , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Imunofluorescência , Humanos , Imuno-Histoquímica , Lactente , Masculino , Camundongos , Camundongos Knockout , Microfibrilas/ultraestrutura , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/imunologia , Microscopia Eletrônica , Dados de Sequência Molecular , Músculo Esquelético/química , Homologia de Sequência de Aminoácidos , Pele/química
20.
J Cell Physiol ; 226(6): 1499-509, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20945348

RESUMO

The latent TGF-ß binding proteins (LTBP-1 -3, and -4) assist in the secretion and localization of latent TGF-ß molecules. Ltbp3(-/-) and Ltbp4S(-/-) mice have distinct phenotypes and only in the lungs does deficiency of either Ltbp-3 or Ltbp-4 cause developmental abnormalities. To determine if these two LTBPs have additional common functions, we generated mice deficient for both Ltbp-3 and Ltbp-4S. The only novel defect in Ltbp3(-/-);Ltbp4S(-/-) mice was an early lethality compared to mice with single mutations. In addition lung abnormalities were exacerbated and the terminal air sac septation defect was more severe in Ltbp3(-/-);Ltbp4S(-/-) mice than in Ltbp4S(-/-) mice. Decreased cellularity of Ltbp3(-/-);Ltbp4S(-/-) lungs was correlated with higher rate of apoptosis in newborn lungs of Ltbp3(-/-);Ltbp4S(-/-) animals compared to WT, Ltbp3(-/-), and Ltbp4S(-/-) mice. No differences in the maturation of the major lung cell types were discerned between the single and double mutant mice. However, the distribution of type 2 cells and myofibroblasts was abnormal, and myofibroblast segregation in some areas might be an indication of early fibrosis. We also observed differences in ECM composition between Ltbp3(-/-);Ltbp4S(-/-) and Ltbp4S(-/-) lungs after birth, reflected in decreased incorporation of fibrillin-1 and -2 in Ltbp3(-/-);Ltbp4S(-/-) matrix. The function of the lungs of Ltbp3(-/-);Ltbp4S(-/-) mice after the first week of life was potentially further compromised by macrophage infiltration, as proteases secreted from macrophages might exacerbate developmental emphysema. Together these data indicate that LTBP-3 and -4 perform partially overlapping functions only in the lungs.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Animais , Apoptose , Diferenciação Celular , Elastina/biossíntese , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inflamação/metabolismo , Inflamação/patologia , Proteínas de Ligação a TGF-beta Latente/deficiência , Proteínas de Ligação a TGF-beta Latente/genética , Pulmão/patologia , Camundongos , Microfibrilas/metabolismo , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA