Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Am J Hum Genet ; 109(12): 2230-2252, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36351433

RESUMEN

EMILIN1 (elastin-microfibril-interface-located-protein-1) is a structural component of the elastic fiber network and localizes to the interface between the fibrillin microfibril scaffold and the elastin core. How EMILIN1 contributes to connective tissue integrity is not fully understood. Here, we report bi-allelic EMILIN1 loss-of-function variants causative for an entity combining cutis laxa, arterial tortuosity, aneurysm formation, and bone fragility, resembling autosomal-recessive cutis laxa type 1B, due to EFEMP2 (FBLN4) deficiency. In both humans and mice, absence of EMILIN1 impairs EFEMP2 extracellular matrix deposition and LOX activity resulting in impaired elastogenesis, reduced collagen crosslinking, and aberrant growth factor signaling. Collagen fiber ultrastructure and histopathology in EMILIN1- or EFEMP2-deficient skin and aorta corroborate these findings and murine Emilin1-/- femora show abnormal trabecular bone formation and strength. Altogether, EMILIN1 connects elastic fiber network with collagen fibril formation, relevant for both bone and vascular tissue homeostasis.


Asunto(s)
Enfermedades Óseas Metabólicas , Cutis Laxo , Animales , Humanos , Ratones , Colágeno/genética , Cutis Laxo/genética , Elastina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo
2.
Circ Res ; 123(6): 660-672, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30355232

RESUMEN

RATIONALE: Abnormal mechanosensing of smooth muscle cells (SMCs) resulting from the defective elastin-contractile units has been suggested to drive the formation of thoracic aortic aneurysms; however, the precise molecular mechanism has not been elucidated. OBJECTIVE: The aim of this study was to identify the crucial mediator(s) involved in abnormal mechanosensing and propagation of biochemical signals during the aneurysm formation and to establish a basis for a novel therapeutic strategy. METHODS AND RESULTS: We used a mouse model of postnatal ascending aortic aneurysms ( Fbln4SMKO; termed SMKO [SMC-specific knockout]), in which deletion of Fbln4 (fibulin-4) leads to disruption of the elastin-contractile units caused by a loss of elastic lamina-SMC connections. In this mouse, upregulation of Egr1 (early growth response 1) and angiotensin-converting enzyme leads to activation of Ang II (angiotensin II) signaling. Here, we showed that the matricellular protein, Thbs1 (thrombospondin-1), was highly upregulated in SMKO ascending aortas and in human thoracic aortic aneurysms. Thbs1 was induced by mechanical stretch and Ang II in SMCs, for which Egr1 was required, and reduction of Fbln4 sensitized the cells to these stimuli and led to higher expression of Egr1 and Thbs1. Deletion of Thbs1 in SMKO mice prevented the aneurysm formation in ≈80% of DKO (SMKO;Thbs1 knockout) animals and suppressed Ssh1 (slingshot-1) and cofilin dephosphorylation, leading to the formation of normal actin filaments. Furthermore, elastic lamina-SMC connections were restored in DKO aortas, and mechanical testing showed that structural and material properties of DKO aortas were markedly improved. CONCLUSIONS: Thbs1 is a critical component of mechanotransduction, as well as a modulator of elastic fiber organization. Maladaptive upregulation of Thbs1 results in disruption of elastin-contractile units and dysregulation of actin cytoskeletal remodeling, contributing to the development of ascending aortic aneurysms in vivo. Thbs1 may serve as a potential therapeutic target for treating thoracic aortic aneurysms.


Asunto(s)
Aneurisma de la Aorta Torácica/metabolismo , Mecanotransducción Celular , Músculo Liso Vascular/metabolismo , Trombospondina 1/metabolismo , Remodelación Vascular , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Anciano , Anciano de 80 o más Años , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/patología , Aneurisma de la Aorta Torácica/prevención & control , Células Cultivadas , Cofilina 2/metabolismo , Dilatación Patológica , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Tejido Elástico/metabolismo , Tejido Elástico/patología , Elastina/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Femenino , Humanos , Masculino , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/patología , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Presorreceptores/metabolismo , Ratas , Estrés Mecánico , Trombospondina 1/deficiencia , Trombospondina 1/genética
3.
Circ Res ; 120(12): 1903-1915, 2017 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-28461455

RESUMEN

RATIONALE: Mutations in ACTA2, encoding the smooth muscle isoform of α-actin, cause thoracic aortic aneurysms, acute aortic dissections, and occlusive vascular diseases. OBJECTIVE: We sought to identify the mechanism by which loss of smooth muscle α-actin causes aortic disease. METHODS AND RESULTS: Acta2-/- mice have an increased number of elastic lamellae in the ascending aorta and progressive aortic root dilation as assessed by echocardiography that can be attenuated by treatment with losartan, an angiotensin II (AngII) type 1 receptor blocker. AngII levels are not increased in Acta2-/- aortas or kidneys. Aortic tissue and explanted smooth muscle cells from Acta2-/- aortas show increased production of reactive oxygen species and increased basal nuclear factor κB signaling, leading to an increase in the expression of the AngII receptor type I a and activation of signaling at 100-fold lower levels of AngII in the mutant compared with wild-type cells. Furthermore, disruption of smooth muscle α-actin filaments in wild-type smooth muscle cells by various mechanisms activates nuclear factor κB signaling and increases expression of AngII receptor type I a. CONCLUSIONS: These findings reveal that disruption of smooth muscle α-actin filaments in smooth muscle cells increases reactive oxygen species levels, activates nuclear factor κB signaling, and increases AngII receptor type I a expression, thus potentiating AngII signaling in vascular smooth muscle cells without an increase in the exogenous levels of AngII.


Asunto(s)
Actinas/deficiencia , Angiotensina II/metabolismo , Aorta Torácica/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Receptor de Angiotensina Tipo 1/biosíntesis , Actinas/efectos de los fármacos , Actinas/genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Células Cultivadas , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/genética
4.
Nature ; 503(7474): 126-30, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24107997

RESUMEN

In systemic sclerosis (SSc), a common and aetiologically mysterious form of scleroderma (defined as pathological fibrosis of the skin), previously healthy adults acquire fibrosis of the skin and viscera in association with autoantibodies. Familial recurrence is extremely rare and causal genes have not been identified. Although the onset of fibrosis in SSc typically correlates with the production of autoantibodies, whether they contribute to disease pathogenesis or simply serve as a marker of disease remains controversial and the mechanism for their induction is largely unknown. The study of SSc is hindered by a lack of animal models that recapitulate the aetiology of this complex disease. To gain a foothold in the pathogenesis of pathological skin fibrosis, we studied stiff skin syndrome (SSS), a rare but tractable Mendelian disorder leading to childhood onset of diffuse skin fibrosis with autosomal dominant inheritance and complete penetrance. We showed previously that SSS is caused by heterozygous missense mutations in the gene (FBN1) encoding fibrillin-1, the main constituent of extracellular microfibrils. SSS mutations all localize to the only domain in fibrillin-1 that harbours an Arg-Gly-Asp (RGD) motif needed to mediate cell-matrix interactions by binding to cell-surface integrins. Here we show that mouse lines harbouring analogous amino acid substitutions in fibrillin-1 recapitulate aggressive skin fibrosis that is prevented by integrin-modulating therapies and reversed by antagonism of the pro-fibrotic cytokine transforming growth factor ß (TGF-ß). Mutant mice show skin infiltration of pro-inflammatory immune cells including plasmacytoid dendritic cells, T helper cells and plasma cells, and also autoantibody production; these findings are normalized by integrin-modulating therapies or TGF-ß antagonism. These results show that alterations in cell-matrix interactions are sufficient to initiate and sustain inflammatory and pro-fibrotic programmes and highlight new therapeutic strategies.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Contractura/tratamiento farmacológico , Contractura/patología , Integrinas/efectos de los fármacos , Integrinas/metabolismo , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Enfermedades Cutáneas Genéticas/tratamiento farmacológico , Enfermedades Cutáneas Genéticas/patología , Secuencias de Aminoácidos/genética , Sustitución de Aminoácidos/genética , Animales , Anticuerpos Antinucleares/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Autoinmunidad/inmunología , Contractura/inmunología , Contractura/prevención & control , Células Dendríticas/efectos de los fármacos , Femenino , Fibrilina-1 , Fibrilinas , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Fibrosis/prevención & control , Masculino , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Mutación Missense/genética , Células Plasmáticas/efectos de los fármacos , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/prevención & control , Enfermedades Cutáneas Genéticas/inmunología , Enfermedades Cutáneas Genéticas/prevención & control , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/inmunología
5.
Hum Mol Genet ; 24(20): 5867-79, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220971

RESUMEN

Homozygous recessive mutations in either EFEMP2 (encoding fibulin-4) or FBLN5 (encoding fibulin-5), critical genes for elastogenesis, lead to autosomal recessive cutis laxa types 1B and 1A, respectively. Previously, fibulin-4 was shown to bind lysyl oxidase (LOX), an elastin/collagen cross-linking enzyme, in vitro. Consistently, reported defects in humans with EFEMP2 mutations are more severe and broad in range than those due to FBLN5 mutations and encompass both elastin-rich and collagen-rich tissues. However, the underlying disease mechanism in EFEMP2 mutations has not been fully addressed. Here, we show that fibulin-4 is important for the integrity of aortic collagen in addition to elastin. Smooth muscle-specific Efemp2 loss in mouse (termed SMKO) resulted in altered fibrillar collagen localization with larger, poorly organized fibrils. LOX activity was decreased in Efemp2-null cells, and collagen cross-linking was diminished in SMKO aortas; however, elastin cross-linking was unaffected and the level of mature LOX was maintained to that of wild-type aortas. Proteomic screening identified multiple proteins involved in procollagen processing and maturation as potential fibulin-4-binding partners. We showed that fibulin-4 binds procollagen C-endopeptidase enhancer 1 (Pcolce), which enhances proteolytic cleavage of the procollagen C-terminal propeptide during procollagen processing. Interestingly, however, procollagen cleavage was not affected by the presence or absence of fibulin-4 in vitro. Thus, our data indicate that fibulin-4 serves as a potential scaffolding protein during collagen maturation in the extracellular space. Analysis of collagen in other tissues affected by fibulin-4 loss should further increase our understanding of underlying pathologic mechanisms in patients with EFEMP2 mutations.


Asunto(s)
Aorta/metabolismo , Colágeno/biosíntesis , Proteínas de la Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Animales , Colágeno/metabolismo , Elastina/metabolismo , Eliminación de Gen , Homocigoto , Ratones , Músculo Liso/metabolismo , Oxidación-Reducción , Proteína-Lisina 6-Oxidasa/metabolismo , Proteómica
6.
Hum Mol Genet ; 24(14): 4024-36, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882708

RESUMEN

Mutations in the gene for the latent transforming growth factor beta binding protein 4 (LTBP4) cause autosomal recessive cutis laxa type 1C. To understand the molecular disease mechanisms of this disease, we investigated the impact of LTBP4 loss on transforming growth factor beta (TGFß) signaling. Despite elevated extracellular TGFß activity, downstream signaling molecules of the TGFß pathway, including pSMAD2 and pERK, were down-regulated in LTBP4 mutant human dermal fibroblasts. In addition, TGFß receptors 1 and 2 (TGFBR1 and TGFBR2) were reduced at the protein but not at the ribonucleic acid level. Treatment with exogenous TGFß1 led to an initially rapid increase in SMAD2 phosphorylation followed by a sustained depression of phosphorylation and receptor abundance. In mutant cells TGFBR1 was co-localized with lysosomes. Treatment with a TGFBR1 kinase inhibitor, endocytosis inhibitors or a lysosome inhibitor, normalized the levels of TGFBR1 and TGFBR2. Co-immunoprecipitation demonstrated a molecular interaction between LTBP4 and TGFBR2. Knockdown of LTBP4 reduced TGFß receptor abundance and signaling in normal cells and supplementation of recombinant LTBP4 enhanced these measures in mutant cells. In a mouse model of Ltbp4 deficiency, reduced TGFß signaling and receptor levels were normalized upon TGFBR1 kinase inhibitor treatment. Our results show that LTBP4 interacts with TGFBR2 and stabilizes TGFß receptors by preventing their endocytosis and lysosomal degradation in a ligand-dependent and receptor kinase activity-dependent manner. These findings identify LTBP4 as a key molecule required for the stability of the TGFß receptor complex, and a new mechanism by which the extracellular matrix regulates cytokine receptor signaling.


Asunto(s)
Cutis Laxo/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Endocitosis/genética , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Inmunoprecipitación , Proteínas de Unión a TGF-beta Latente/genética , Masculino , Ratones , Ratones Noqueados , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo
7.
J Cell Physiol ; 230(1): 226-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24962333

RESUMEN

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.


Asunto(s)
Tipificación del Cuerpo/genética , Tejido Elástico/embriología , Proteínas de la Matriz Extracelular/fisiología , Proteínas de Unión a TGF-beta Latente/fisiología , Pulmón/embriología , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Tejido Elástico/anomalías , Elastina/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibrilinas , Proteínas de Unión a TGF-beta Latente/genética , Pulmón/anomalías , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteínas Recombinantes/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta2/genética
8.
Hum Mol Genet ; 22(1): 1-17, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22949511

RESUMEN

Although biallelic mutations in non-collagen genes account for <10% of individuals with osteogenesis imperfecta, the characterization of these genes has identified new pathways and potential interventions that could benefit even those with mutations in type I collagen genes. We identified mutations in FKBP10, which encodes the 65 kDa prolyl cis-trans isomerase, FKBP65, in 38 members of 21 families with OI. These include 10 families from the Samoan Islands who share a founder mutation. Of the mutations, three are missense; the remainder either introduce premature termination codons or create frameshifts both of which result in mRNA instability. In four families missense mutations result in loss of most of the protein. The clinical effects of these mutations are short stature, a high incidence of joint contractures at birth and progressive scoliosis and fractures, but there is remarkable variability in phenotype even within families. The loss of the activity of FKBP65 has several effects: type I procollagen secretion is slightly delayed, the stabilization of the intact trimer is incomplete and there is diminished hydroxylation of the telopeptide lysyl residues involved in intermolecular cross-link formation in bone. The phenotype overlaps with that seen with mutations in PLOD2 (Bruck syndrome II), which encodes LH2, the enzyme that hydroxylates the telopeptide lysyl residues. These findings define a set of genes, FKBP10, PLOD2 and SERPINH1, that act during procollagen maturation to contribute to molecular stability and post-translational modification of type I procollagen, without which bone mass and quality are abnormal and fractures and contractures result.


Asunto(s)
Artrogriposis/genética , Colágeno Tipo I/metabolismo , Genes Recesivos , Lisina/metabolismo , Mutación , Osteogénesis Imperfecta/genética , Proteínas de Unión a Tacrolimus/genética , Femenino , Humanos , Hidroxilación , Masculino , Procesamiento Proteico-Postraduccional
9.
Am J Physiol Regul Integr Comp Physiol ; 308(7): R576-89, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25608750

RESUMEN

Activation of muscle progenitor cell myogenesis and endothelial cell angiogenesis is critical for the recovery of skeletal muscle from injury. Angiopoietin-1 (Ang-1), a ligand of Tie-2 receptors, enhances angiogenesis and skeletal muscle satellite cell survival; however, its role in skeletal muscle regeneration after injury is unknown. We assessed the effects of Ang-1 on fiber regeneration, myogenesis, and angiogenesis in injured skeletal muscle (tibialis anterior, TA) in mice. We also assessed endogenous Ang-1 levels and localization in intact and injured TA muscles. TA fiber injury was triggered by cardiotoxin injection. Endogenous Ang-1 mRNA levels immediately decreased in response to cardiotoxin then increased during the 2 wk. Ang-1 protein was expressed in satellite cells, both in noninjured and recovering TA muscles. Positive Ang-1 staining was present in blood vessels but not in nerve fibers. Four days after the initiation of injury, injection of adenoviral Ang-1 into injured muscles resulted in significant increases in in situ TA muscle contractility, muscle fiber regeneration, and capillary density. In cultured human skeletal myoblasts, recombinant Ang-1 protein increased survival, proliferation, migration, and differentiation into myotubes. The latter effect was associated with significant upregulation of the expression of the myogenic regulatory factors MyoD and Myogenin and certain genes involved in cell cycle regulation. We conclude that Ang-1 strongly enhances skeletal muscle regeneration in response to fiber injury and that this effect is mediated through induction of the myogenesis program in muscle progenitor cells and the angiogenesis program in endothelial cells.


Asunto(s)
Angiopoyetina 1/metabolismo , Terapia Genética/métodos , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Enfermedades Musculares/terapia , Regeneración , Adenoviridae/genética , Adulto , Angiopoyetina 1/genética , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Cardiotoxinas , Diferenciación Celular , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Mioblastos/metabolismo , Mioblastos/patología , Necrosis , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo
10.
Hum Mol Genet ; 21(6): 1248-59, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22116938

RESUMEN

Growth factor signaling results in dramatic phenotypic changes in cells, which require commensurate alterations in cellular metabolism. Mutations in SLC2A10/GLUT10, a member of the facilitative glucose transporter family, are associated with altered transforming growth factor-ß (TGFß) signaling in patients with arterial tortuosity syndrome (ATS). The objective of this work was to test whether SLC2A10/GLUT10 can serve as a link between TGFß-related transcriptional regulation and metabolism during development. In zebrafish embryos, knockdown of slc2a10 using antisense morpholino oligonucleotide injection caused a wavy notochord and cardiovascular abnormalities with a reduced heart rate and blood flow, which was coupled with an incomplete and irregular vascular patterning. This was phenocopied by treatment with a small-molecule inhibitor of TGFß receptor (tgfbr1/alk5). Array hybridization showed that the changes at the transcriptome level caused by the two treatments were highly correlated, revealing that a reduced tgfbr1 signaling is a key feature of ATS in early zebrafish development. Interestingly, a large proportion of the genes, which were specifically dysregulated after glut10 depletion gene and not by tgfbr1 inhibition, play a major role in mitochondrial function. Consistent with these results, slc2a10 morphants showed decreased respiration and reduced TGFß reporter gene activity. Finally, co-injection of antisense morpholinos targeting slc2a10 and smad7 (a TGFß inhibitor) resulted in a partial rescue of smad7 morphant phenotypes, suggesting scl2a10/glut10 functions downstream of smads. Taken together, glut10 is essential for cardiovascular development by facilitating both mitochondrial respiration and TGFß signaling.


Asunto(s)
Anomalías Cardiovasculares/etiología , Proteínas Facilitadoras del Transporte de la Glucosa/fisiología , Mitocondrias/metabolismo , Notocorda/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/patología , Luciferasas/metabolismo , Mitocondrias/patología , Datos de Secuencia Molecular , Morfolinos/farmacología , Mutación/genética , Notocorda/patología , Fenotipo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Transcriptoma , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Pez Cebra/crecimiento & desarrollo
11.
Nat Genet ; 37(3): 275-81, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15731757

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo I/genética , Desarrollo Óseo/genética , Sistema Cardiovascular/crecimiento & desarrollo , Trastornos del Conocimiento/genética , Cara , Mutación , Receptores de Factores de Crecimiento Transformadores beta/genética , Cráneo/crecimiento & desarrollo , Secuencia de Aminoácidos , Preescolar , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Fenotipo , Proteínas Serina-Treonina Quinasas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Homología de Secuencia de Aminoácido , Síndrome
12.
Hum Mutat ; 34(1): 111-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22829427

RESUMEN

Autosomal recessive cutis laxa type I (ARCL type I) is characterized by generalized cutis laxa with pulmonary emphysema and/or vascular complications. Rarely, mutations can be identified in FBLN4 or FBLN5. Recently, LTBP4 mutations have been implicated in a similar phenotype. Studying FBLN4, FBLN5, and LTBP4 in 12 families with ARCL type I, we found bi-allelic FBLN5 mutations in two probands, whereas nine probands harbored biallelic mutations in LTBP4. FBLN5 and LTBP4 mutations cause a very similar phenotype associated with severe pulmonary emphysema, in the absence of vascular tortuosity or aneurysms. Gastrointestinal and genitourinary tract involvement seems to be more severe in patients with LTBP4 mutations. Functional studies showed that most premature termination mutations in LTBP4 result in severely reduced mRNA and protein levels. This correlated with increased transforming growth factor-beta (TGFß) activity. However, one mutation, c.4127dupC, escaped nonsense-mediated decay. The corresponding mutant protein (p.Arg1377Alafs(*) 27) showed reduced colocalization with fibronectin, leading to an abnormal morphology of microfibrils in fibroblast cultures, while retaining normal TGFß activity. We conclude that LTBP4 mutations cause disease through both loss of function and gain of function mechanisms.


Asunto(s)
Cutis Laxo/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de Unión a TGF-beta Latente/genética , Mutación , Adolescente , Secuencia de Bases , Western Blotting , Niño , Preescolar , Consanguinidad , Cutis Laxo/complicaciones , Proteínas de la Matriz Extracelular/metabolismo , Salud de la Familia , Femenino , Expresión Génica , Humanos , Lactante , Proteínas de Unión a TGF-beta Latente/metabolismo , Masculino , Microscopía Electrónica , Linaje , Enfisema Pulmonar/complicaciones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Piel/metabolismo , Piel/patología , Piel/ultraestructura , Adulto Joven
13.
J Biol Chem ; 287(51): 43180-90, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23105101

RESUMEN

Protein-tyrosine phosphatase (PTP)-PEST (PTPN12) is ubiquitously expressed. It is essential for normal embryonic development and embryonic viability in mice. Herein we addressed the involvement of PTP-PEST in endothelial cell functions using a combination of genetic and biochemical approaches. By generating primary endothelial cells from an inducible PTP-PEST-deficient mouse, we found that PTP-PEST is not needed for endothelial cell differentiation and proliferation or for the control of endothelial cell permeability. Nevertheless, it is required for integrin-mediated adhesion and migration of endothelial cells. PTP-PEST-deficient endothelial cells displayed increased tyrosine phosphorylation of Cas, paxillin, and Pyk2, which were previously also implicated in integrin functions. By eliminating PTP-PEST in endothelial cells in vivo, we obtained evidence that expression of PTP-PEST in endothelial cells is required for normal vascular development and embryonic viability. Therefore, PTP-PEST is a key regulator of integrin-mediated functions in endothelial cells seemingly through its capacity to control Cas, paxillin, and Pyk2. This function explains at least in part the essential role of PTP-PEST in embryonic development and viability.


Asunto(s)
Vasos Sanguíneos/embriología , Permeabilidad de la Membrana Celular , Movimiento Celular , Embrión de Mamíferos/enzimología , Células Endoteliales/citología , Células Endoteliales/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Animales , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Embrión de Mamíferos/citología , Femenino , Quinasa 2 de Adhesión Focal/metabolismo , Genotipo , Integrinas/metabolismo , Uniones Intercelulares/metabolismo , Ratones , Ratones Endogámicos C57BL , Paxillin/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 12/deficiencia
14.
J Biol Chem ; 287(26): 22055-67, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22573328

RESUMEN

Elastin is the extracellular matrix protein in vertebrates that provides elastic recoil to blood vessels, the lung, and skin. Because the elastin gene has undergone significant changes in the primate lineage, modeling elastin diseases in non-human animals can be problematic. To investigate the pathophysiology underlying a class of elastin gene mutations leading to autosomal dominant cutis laxa, we engineered a cutis laxa mutation (single base deletion) into the human elastin gene contained in a bacterial artificial chromosome. When expressed as a transgene in mice, mutant elastin was incorporated into elastic fibers in the skin and lung with adverse effects on tissue function. In contrast, only low levels of mutant protein incorporated into aortic elastin, which explains why the vasculature is relatively unaffected in this disease. RNA stability studies found that alternative exon splicing acts as a modifier of disease severity by influencing the spectrum of mutant transcripts that survive nonsense-mediated decay. Our results confirm the critical role of the C-terminal region of tropoelastin in elastic fiber assembly and suggest tissue-specific differences in the elastin assembly pathway.


Asunto(s)
Empalme Alternativo , Cutis Laxo/genética , Elastina/biosíntesis , Elastina/genética , Mutación , Animales , Aorta/metabolismo , Cromosomas Artificiales Bacterianos , Reactivos de Enlaces Cruzados/química , Elasticidad , Elastina/metabolismo , Exones , Fibroblastos/citología , Mutación del Sistema de Lectura , Genes Dominantes , Humanos , Ratones , Ratones Transgénicos , Estructura Terciaria de Proteína , ARN/química , Transgenes
15.
Am J Hum Genet ; 86(4): 551-9, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20362275

RESUMEN

Osteogenesis imperfecta is a clinically and genetically heterogeneous brittle bone disorder that results from defects in the synthesis, structure, or posttranslational modification of type I procollagen. Dominant forms of OI result from mutations in COL1A1 or COL1A2, which encode the chains of the type I procollagen heterotrimer. The mildest form of OI typically results from diminished synthesis of structurally normal type I procollagen, whereas moderately severe to lethal forms of OI usually result from structural defects in one of the type I procollagen chains. Recessively inherited OI, usually phenotypically severe, has recently been shown to result from defects in the prolyl-3-hydroxylase complex that lead to the absence of a single 3-hydroxyproline at residue 986 of the alpha1(I) triple helical domain. We studied a cohort of five consanguineous Turkish families, originating from the Black Sea region of Turkey, with moderately severe recessively inherited OI and identified a novel locus for OI on chromosome 17. In these families, and in a Mexican-American family, homozygosity for mutations in FKBP10, which encodes FKBP65, a chaperone that participates in type I procollagen folding, was identified. Further, we determined that FKBP10 mutations affect type I procollagen secretion. These findings identify a previously unrecognized mechanism in the pathogenesis of OI.


Asunto(s)
Genes Recesivos , Mutación/genética , Osteogénesis Imperfecta/genética , Proteínas de Unión a Tacrolimus/genética , Adolescente , Estudios de Casos y Controles , Niño , Estudios de Cohortes , Colágeno Tipo I/genética , Femenino , Homocigoto , Humanos , Masculino , Osteogénesis Imperfecta/patología , Linaje , Fenotipo , Piel/patología
16.
Respir Res ; 14: 110, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24138138

RESUMEN

BACKGROUND: Airway inflammation and airway remodeling are the key contributors to airway hyperresponsiveness (AHR), a characteristic feature of asthma. Both processes are regulated by Transforming Growth Factor (TGF)-ß. Caveolin 1 (Cav1) is a membrane bound protein that binds to a variety of receptor and signaling proteins, including the TGF-ß receptors. We hypothesized that caveolin-1 deficiency promotes structural alterations of the airways that develop with age will predispose to an increased response to allergen challenge. METHODS: AHR was measured in Cav1-deficient and wild-type (WT) mice 1 to 12 months of age to examine the role of Cav1 in AHR and the relative contribution of inflammation and airway remodeling. AHR was then measured in Cav1-/- and WT mice after an ovalbumin-allergen challenge performed at either 2 months of age, when remodeling in Cav1-/- and WT mice was equivalent, and at 6 months of age, when the Cav1-/- mice had established airway remodeling. RESULTS: Cav1-/- mice developed increased thickness of the subepithelial layer and a correspondingly increased AHR as they aged. In addition, allergen-challenged Cav1-/- mice had an increase in AHR greater than WT mice that was largely independent of inflammation. Cav1-/- mice challenged at 6 months of age have decreased AHR compared to those challenged at 2 months with correspondingly decreased BAL IL-4 and IL-5 levels, inflammatory cell counts and percentage of eosinophils. In addition, in response to OVA challenge, the number of goblet cells and α-SMA positive cells in the airways were reduced with age in response to OVA challenge in contrast to an increased collagen deposition further enhanced in absence of Cav1. CONCLUSION: A lack of Cav1 contributed to the thickness of the subepithelial layer in mice as they aged resulting in an increase in AHR independent of inflammation, demonstrating the important contribution of airway structural changes to AHR. In addition, age in the Cav1-/- mice is a contributing factor to airway remodeling in the response to allergen challenge.


Asunto(s)
Envejecimiento/fisiología , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/fisiopatología , Hiperreactividad Bronquial/fisiopatología , Caveolina 1/deficiencia , Neumonía/fisiopatología , Actinas/metabolismo , Animales , Asma/inducido químicamente , Hiperreactividad Bronquial/inducido químicamente , Caveolina 1/genética , Caveolina 1/fisiología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/efectos adversos , Factor de Crecimiento Transformador beta/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 32(12): 2901-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23042818

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate potential mechanisms promoting abdominal aortic aneurysm development with tobacco smoke (TS) exposure. METHODS AND RESULTS: Experiments used the elastase perfusion model of abdominal aortic aneurysms with smoke-free controls. The effect of TS exposure was evaluated in C57/Bl6 mice, after broad-spectrum matrix metalloproteinase inhibition with doxycycline and in mice deficient in matrix metalloproteinase-9, matrix metalloproteinase-12, Cathepsin-S, and Neutrophil Elastase. Preparations of washed marrow, spleen, and peripheral blood leukocytes were transferred to smoke-free mice from 6-week TS-exposed mice or smoke-free mice. All mice were euthanized 14 days after elastase perfusion, and the percentage of change in aortic diameter (%Δ aortic diameter) was calculated. Electron microscopy of aortic tissue from animals exposed to TS without elastase exposure did not demonstrate any ultrastructural changes. Neither doxycycline nor any specific elastase deficiency was effective at preventing an increase in %Δ aortic diameter in TS-exposed animals. Smoke exposure for 6 weeks increased the %Δ aortic diameter after a smoke-free interval of up to 6 weeks before elastase perfusion. Leukocyte preparations from TS-exposed mice localized to abdominal aortic aneurysms and increased the %Δ aortic diameter in smoke-free mice. CONCLUSIONS: The effect of TS on the development of abdominal aortic aneurysms is not dependent on the activity of elastolytic enzymes and persists for long periods despite cessation of TS. Alterations in leukocyte response to aortic injury appear to mediate this effect.


Asunto(s)
Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/fisiopatología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/fisiología , Fumar/efectos adversos , Animales , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Aorta Abdominal/ultraestructura , Aneurisma de la Aorta Abdominal/patología , Catepsinas/deficiencia , Catepsinas/genética , Catepsinas/fisiología , Recuento de Células , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Elastasa de Leucocito/fisiología , Masculino , Metaloproteinasa 12 de la Matriz/deficiencia , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/fisiología , Metaloproteinasas de la Matriz/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Arterioscler Thromb Vasc Biol ; 32(1): e1-11, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21979435

RESUMEN

OBJECTIVE: Transforming growth factor-ß (TGF-ß) signaling is required for normal vascular development. We aimed to discover the role of TGF-ß signaling in embryonic smooth muscle cells (SMCs). METHODS AND RESULTS: We bred mice with smooth muscle (SM) 22α-Cre and Tgfbr2(flox) alleles to generate embryos in which the type II TGF-ß receptor (TGFBR2; required for TGF-ß signaling) was deleted in SMCs. Embryos were harvested between embryonic day (E) 9.5 and E18.5 and examined grossly, microscopically, and by histochemical and RNA analyses. SM22α-Cre(+/0) Tgfbr2(flox/flox) (knockout [KO]) embryos died before E15.5 with defects that included cardiac outflow tract abnormalities, persistence of the right dorsal aorta, and dilation of the distal aorta. Histological analyses suggested normal expression of SMC differentiation markers in KO aortas; however, RNA analyses showed that SMC differentiation markers were increased in KO cardiac outflow vessels but decreased in the descending aorta. KO aortas had only rare mature elastin deposits and contained abnormal aggregates of extracellular matrix proteins. Expression of several matrix proteins was significantly decreased in KO descending aortas but not in cardiac outflow vessels. CONCLUSIONS: TGF-ß signaling in SMCs controls differentiation, matrix synthesis, and vascular morphogenesis. Effects of TGF-ß on SMC gene expression appear to differ depending on the location of SMCs in the aorta.


Asunto(s)
Mioblastos del Músculo Liso/citología , Mioblastos del Músculo Liso/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Aorta/anomalías , Aorta/embriología , Aorta/metabolismo , Vasos Sanguíneos/embriología , Diferenciación Celular/fisiología , Proteínas de la Matriz Extracelular/biosíntesis , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/fisiología , Neovascularización Fisiológica , Embarazo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal
19.
Am J Hum Genet ; 85(5): 593-605, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19836010

RESUMEN

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.


Asunto(s)
Dermis/anomalías , Intestinos/anomalías , Proteínas de Unión a TGF-beta Latente/genética , Pulmón/anomalías , Mutación , Sistema Urinario/anomalías , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , ADN/genética , ADN/aislamiento & purificación , Dermis/metabolismo , Dermis/ultraestructura , Femenino , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Homocigoto , Humanos , Inmunohistoquímica , Lactante , Mucosa Intestinal/metabolismo , Proteínas de Unión a TGF-beta Latente/química , Pulmón/metabolismo , Masculino , Sistema Musculoesquelético , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Piel/citología , Síndrome , Sistema Urinario/metabolismo
20.
J Cell Sci ; 123(Pt 24): 4221-30, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21081647

RESUMEN

Carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1) is an immunoglobulin-like cell surface co-receptor expressed on epithelial, hematopoietic and endothelial cells. CEACAM1 functions as an adhesion molecule, mainly binding to itself or other members of the CEA family. We and others have previously shown that CEACAM1 is crucial for in vivo vascular integrity during ischemic neo-vascularization. Here, we have deciphered the roles of CEACAM1 in normal and pathological vascularization. We have found that Ceacam1-/- mice exhibit a significant increase in basal vascular permeability related to increased basal Akt and endothelial nitric oxide synthase (eNOS) activation in primary murine lung endothelial cells (MLECs). Moreover, CEACAM1 deletion in MLECs inhibits VEGF-mediated nitric oxide (NO) production, consistent with defective VEGF-dependent in vivo permeability in Ceacam1-/- mice. In addition, Ceacam1-null mice exhibit increased permeability of tumor vasculature. Finally, we demonstrate that CEACAM1 is tyrosine-phosphorylated upon VEGF treatment in a SHP-1- and Src-dependent manner, and that the key residues of the long cytoplasmic domain of CEACAM1 are crucial for CEACAM1 phosphorylation and NO production. This data represents the first report, to our knowledge, of a functional link between CEACAM1 and the VEGFR2/Akt/eNOS-mediated vascular permeability pathway.


Asunto(s)
Permeabilidad Capilar , Antígeno Carcinoembrionario/metabolismo , Secuencias de Aminoácidos , Animales , Aorta/efectos de los fármacos , Aorta/patología , Aorta/ultraestructura , Permeabilidad Capilar/efectos de los fármacos , Antígeno Carcinoembrionario/química , Bovinos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Pulmón/citología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Ratones , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda