Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Dyn ; 253(2): 233-254, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37688792

RESUMO

BACKGROUND: Latent TGFß binding protein-2 (LTBP2) is a fibrillin 1 binding component of the microfibril. LTBP2 is the only LTBP protein that does not bind any isoforms of TGFß, although it may interfere with the function of other LTBPs or interact with other signaling pathways. RESULTS: Here, we investigate mice lacking Ltbp2 (Ltbp2-/- ) and identify multiple phenotypes that impact bodyweight and fat mass, and affect bone and skin development. The alterations in skin and bone development are particularly noteworthy since the strength of these tissues is differentially affected by loss of Ltbp2. Interestingly, some tissues that express high levels of Ltbp2, such as the aorta and lung, do not have a developmental or homeostatic phenotype. CONCLUSIONS: Analysis of these mice show that LTBP2 has complex effects on development through direct effects on the extracellular matrix (ECM) or on signaling pathways that are known to regulate the ECM.


Assuntos
Proteínas de Transporte , Matriz Extracelular , Animais , Camundongos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Matriz Extracelular/metabolismo , Fenótipo , Fator de Crescimento Transformador beta/metabolismo , Isoformas de Proteínas/metabolismo , Ligação Proteica
2.
Matrix Biol Plus ; 12: 100085, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34693248

RESUMO

Arterial stiffening is a significant predictor of cardiovascular disease development and mortality. In elastic arteries, stiffening refers to the loss and fragmentation of elastic fibers, with a progressive increase in collagen fibers. Type VIII collagen (Col-8) is highly expressed developmentally, and then once again dramatically upregulated in aged and diseased vessels characterized by arterial stiffening. Yet its biophysical impact on the vessel wall remains unknown. The purpose of this study was to test the hypothesis that Col-8 functions as a matrix scaffold to maintain vessel integrity during extracellular matrix (ECM) development. These changes are predicted to persist into the adult vasculature, and we have tested this in our investigation. Through our in vivo and in vitro studies, we have determined a novel interaction between Col-8 and elastin. Mice deficient in Col-8 (Col8-/-) had reduced baseline blood pressure and increased arterial compliance, indicating an enhanced Windkessel effect in conducting arteries. Differences in both the ECM composition and VSMC activity resulted in Col8-/- carotid arteries that displayed increased crosslinked elastin and functional distensibility, but enhanced catecholamine-induced VSMC contractility. In vitro studies revealed that the absence of Col-8 dramatically increased tropoelastin mRNA and elastic fiber deposition in the ECM, which was decreased with exogenous Col-8 treatment. These findings suggest a causative role for Col-8 in reducing mRNA levels of tropoelastin and the presence of elastic fibers in the matrix. Moreover, we also found that Col-8 and elastin have opposing effects on VSMC phenotype, the former promoting a synthetic phenotype, whereas the latter confers quiescence. These studies further our understanding of Col-8 function and open a promising new area of investigation related to elastin biology.

3.
Function (Oxf) ; 1(1): zqaa004, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32865539

RESUMO

Dramatic cardiomegaly arising from gain-of-function (GoF) mutations in the ATP-sensitive potassium (KATP) channels genes, ABCC9 and KCNJ8, is a characteristic feature of Cantú syndrome (CS). How potassium channel over-activity results in cardiac hypertrophy, as well as the long-term consequences of cardiovascular remodeling in CS, is unknown. Using genome-edited mouse models of CS, we therefore sought to dissect the pathophysiological mechanisms linking KATP channel GoF to cardiac remodeling. We demonstrate that chronic reduction of systemic vascular resistance in CS is accompanied by elevated renin-angiotensin signaling, which drives cardiac enlargement and blood volume expansion. Cardiac enlargement in CS results in elevation of basal cardiac output, which is preserved in aging. However, the cardiac remodeling includes altered gene expression patterns that are associated with pathological hypertrophy and are accompanied by decreased exercise tolerance, suggestive of reduced cardiac reserve. Our results identify a high-output cardiac hypertrophy phenotype in CS which is etiologically and mechanistically distinct from other myocardial hypertrophies, and which exhibits key features of high-output heart failure (HOHF). We propose that CS is a genetically-defined HOHF disorder and that decreased vascular smooth muscle excitability is a novel mechanism for HOHF pathogenesis.


Assuntos
Mutação com Ganho de Função , Canais KATP , Camundongos , Animais , Canais KATP/genética , Mutação com Ganho de Função/genética , Remodelação Ventricular , Receptores de Sulfonilureias/genética , Cardiomegalia/genética , Trifosfato de Adenosina
4.
J Biol Chem ; 295(9): 2687-2697, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31988245

RESUMO

Microfibril-associated glycoprotein-1 (MAGP-1) is a component of vertebrate extracellular matrix (ECM) microfibrils that, together with the fibrillins, contributes to microfibril function. Many of the phenotypes associated with MAGP-1 gene inactivation are consistent with dysregulation of the transforming growth factor ß (TGFß)/bone morphogenetic protein (BMP) signaling system. We have previously shown that full-length MAGP-1 binds active TGFß-1 and some BMPs. The work presented here further defines the growth factor-binding domain of MAGP-1. Using recombinant domains and synthetic peptides, along with surface plasmon resonance analysis to measure the kinetics of the MAGP-1-TGFß-1 interaction, we localized the TGFß- and BMP-binding site in MAGP-1 to a 19-amino acid-long, highly acidic sequence near the N terminus. This domain was specific for binding active, but not latent, TGFß-1. Growth factor activity experiments revealed that TGFß-1 retains signaling activity when complexed with MAGP-1. Furthermore, when bound to fibrillin, MAGP-1 retained the ability to interact with TGFß-1, and active TGFß-1 did not bind fibrillin in the absence of MAGP-1. The absence of MAGP was sufficient to raise the amount of total TGFß stored in the ECM of cultured cells, suggesting that the MAGPs compete with the TGFß large latent complex for binding to microfibrils. Together, these results indicate that MAGP-1 plays an active role in TGFß signaling in the ECM.


Assuntos
Fatores de Processamento de RNA/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrilina-1/metabolismo , Humanos , Ligação Proteica , Transdução de Sinais
5.
JCI Insight ; 52019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31211696

RESUMO

Heterozygous missense mutations in lysyl oxidase (LOX) are associated with thoracic aortic aneurysms and dissections. To assess how LOX mutations modify protein function and lead to aortic disease, we studied the factors that influence the onset and progression of vascular aneurysms in mice bearing a Lox mutation (p.M292R) linked to aortic dilation in humans. We show that mice heterozygous for the M292R mutation did not develop aneurysmal disease unless challenged with increased hemodynamic stress. Vessel dilation was confined to the ascending aorta although both the ascending and descending aortae showed changes in vessel wall structure, smooth muscle cell number and inflammatory cell recruitment that differed between wild-type and mutant animals. Studies with isolated cells found that M292R-mutant Lox is retained in the endoplasmic reticulum and ultimately cleared through an autophagy/proteasome pathway. Because the mutant protein does not transit to the Golgi where copper incorporation occurs, the protein is never catalytically active. These studies show that the M292R mutation results in LOX loss-of-function due to a secretion defect that predisposes the ascending aorta in mice (and by extension humans with similar mutations) to arterial dilation when exposed to risk factors that impart stress to the arterial wall.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Proteínas da Matriz Extracelular/genética , Hipertensão/complicações , Proteína-Lisina 6-Oxidase/genética , Vasodilatação/fisiologia , Dissecção Aórtica/patologia , Dissecção Aórtica/fisiopatologia , Animais , Aorta/citologia , Aorta/patologia , Aorta/fisiopatologia , Aneurisma da Aorta Torácica/patologia , Aneurisma da Aorta Torácica/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Retículo Endoplasmático/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/ultraestrutura , Técnicas de Introdução de Genes , Predisposição Genética para Doença , Complexo de Golgi/metabolismo , Heterozigoto , Humanos , Hipertensão/fisiopatologia , Mutação com Perda de Função , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiopatologia , Músculo Liso Vascular/ultraestrutura , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Proteína-Lisina 6-Oxidase/metabolismo , Fatores de Risco , Estresse Fisiológico
6.
Cell Metab ; 29(2): 475-487.e7, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30415924

RESUMO

Lipoproteins trapped in arteries drive atherosclerosis. Extravascular low-density lipoprotein undergoes receptor uptake, whereas high-density lipoprotein (HDL) interacts with cells to acquire cholesterol and then recirculates to plasma. We developed photoactivatable apoA-I to understand how HDL passage through tissue is regulated. We focused on skin and arteries of healthy mice versus those with psoriasis, which carries cardiovascular risk in man. Our findings suggest that psoriasis-affected skin lesions program interleukin-17-producing T cells in draining lymph nodes to home to distal skin and later to arteries. There, these cells mediate thickening of the collagenous matrix, such that larger molecules including lipoproteins become entrapped. HDL transit was rescued by depleting CD4+ T cells, neutralizing interleukin-17, or inhibiting lysyl oxidase that crosslinks collagen. Experimental psoriasis also increased vascular stiffness and atherosclerosis via this common pathway. Thus, interleukin-17 can reduce lipoprotein trafficking and increase vascular stiffness by, at least in part, remodeling collagen.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Interleucina-17/metabolismo , Lipoproteínas HDL/metabolismo , Psoríase/metabolismo , Pele/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Aterosclerose/metabolismo , Transporte Biológico , Linfócitos T CD4-Positivos/citologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína-Lisina 6-Oxidase/metabolismo , Pele/patologia
7.
Sci Signal ; 11(546)2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30181242

RESUMO

Constitutively active G protein α subunits cause cancer, cholera, Sturge-Weber syndrome, and other disorders. Therapeutic intervention by targeted inhibition of constitutively active Gα subunits in these disorders has yet to be achieved. We found that constitutively active Gαq in uveal melanoma (UM) cells was inhibited by the cyclic depsipeptide FR900359 (FR). FR allosterically inhibited guanosine diphosphate-for-guanosine triphosphate (GDP/GTP) exchange to trap constitutively active Gαq in inactive, GDP-bound Gαßγ heterotrimers. Allosteric inhibition of other Gα subunits was achieved by the introduction of an FR-binding site. In UM cells driven by constitutively active Gαq, FR inhibited second messenger signaling, arrested cell proliferation, reinstated melanocytic differentiation, and stimulated apoptosis. In contrast, FR had no effect on BRAF-driven UM cells. FR promoted UM cell differentiation by reactivating polycomb repressive complex 2 (PRC2)-mediated gene silencing, a heretofore unrecognized effector system of constitutively active Gαq in UM. Constitutively active Gαq and PRC2 therefore provide therapeutic targets for UM. The development of FR analogs specific for other Gα subunit subtypes may provide novel therapeutic approaches for diseases driven by constitutively active Gα subunits or multiple G protein-coupled receptors (GPCRs) where targeting a single receptor is ineffective.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Neoplasias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Depsipeptídeos/farmacologia , Subunidades alfa de Proteínas de Ligação ao GTP/antagonistas & inibidores , Células HEK293 , Humanos , Camundongos , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos
8.
Am J Physiol Heart Circ Physiol ; 315(1): H18-H32, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29498532

RESUMO

Increased vascular stiffness correlates with a higher risk of cardiovascular complications in aging adults. Elastin (ELN) insufficiency, as observed in patients with Williams-Beuren syndrome or with familial supravalvular aortic stenosis, also increases vascular stiffness and leads to arterial narrowing. We used Eln+/- mice to test the hypothesis that pathologically increased vascular stiffness with concomitant arterial narrowing leads to decreased blood flow to end organs such as the brain. We also hypothesized that drugs that remodel arteries and increase lumen diameter would improve flow. To test these hypotheses, we compared carotid blood flow using ultrasound and cerebral blood flow using MRI-based arterial spin labeling in wild-type (WT) and Eln+/- mice. We then studied how minoxidil, an ATP-sensitive K+ channel opener and vasodilator, affects vessel mechanics, blood flow, and gene expression. Both carotid and cerebral blood flows were lower in Eln+/- mice than in WT mice. Treatment of Eln+/- mice with minoxidil lowered blood pressure and reduced functional arterial stiffness to WT levels. Minoxidil also improved arterial diameter and restored carotid and cerebral blood flows in Eln+/- mice. The beneficial effects persisted for weeks after drug removal. RNA-Seq analysis revealed differential expression of 127 extracellular matrix-related genes among the treatment groups. These results indicate that ELN insufficiency impairs end-organ perfusion, which may contribute to the increased cardiovascular risk. Minoxidil, despite lowering blood pressure, improves end-organ perfusion. Changes in matrix gene expression and persistence of treatment effects after drug withdrawal suggest arterial remodeling. Such remodeling may benefit patients with genetic or age-dependent ELN insufficiency. NEW & NOTEWORTHY Our work with a model of chronic vascular stiffness, the elastin ( Eln)+/- mouse, shows reduced brain perfusion as measured by carotid ultrasound and MRI arterial spin labeling. Vessel caliber, functional stiffness, and blood flow improved with minoxidil. The ATP-sensitive K+ channel opener increased Eln gene expression and altered 126 other matrix-associated genes.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Minoxidil/farmacologia , Rigidez Vascular/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiologia , Elastina/genética , Elastina/metabolismo , Matriz Extracelular/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
9.
Matrix Biol ; 71-72: 100-111, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29524629

RESUMO

Microfibril-associated glycoproteins 1 and 2 (MAGP-1, MAGP-2) are protein components of extracellular matrix microfibrils. These proteins interact with fibrillin, the core component of microfibrils, and impart unique biological properties that influence microfibril function in vertebrates. MAGPs bind active forms of TGFß and BMPs and are capable of modulating Notch signaling. Mutations in MAGP-1 or MAGP-2 have been linked to thoracic aneurysms and metabolic disease in humans. MAGP-2 has also been shown to be an important biomarker in several human cancers. Mice lacking MAGP-1 or MAGP-2 have defects in multiple organ systems, which reflects the widespread distribution of microfibrils in vertebrate tissues. This review summarizes our current understanding of the function of the MAGPs and their relationship to human disease.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Biomarcadores/metabolismo , Matriz Extracelular/metabolismo , Humanos , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Camundongos , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Fatores de Processamento de RNA , Receptores Notch/metabolismo , Transdução de Sinais
10.
Methods Cell Biol ; 143: 133-146, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29310774

RESUMO

Elastin and collagen levels in tissues are frequently difficult to measure because of each protein's limited solubility. This chapter provides detailed methodology for the determination of elastin, collagen, and total protein levels in a single tissue sample. All three assays start with an acid hydrolysate of the tissue, which breaks the tissue-associated proteins down to their component amino acids. Marker amino acids unique to each protein (desmosine for elastin and hydroxyproline for collagen) are then quantified. Total protein content, useful as a denominator for data normalization, can also be measured from a portion of the hydrolysate using an assay for free amino groups. These measurements are performed using convenient 96-well assay plates and require only a plate reader to determine absorbance.


Assuntos
Colágeno/análise , Elastina/análise , Animais , Desmosina/química , Elastina/química , Hidrólise , Hidroxiprolina/química , Solubilidade
11.
Sci Adv ; 3(5): e1602532, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28508064

RESUMO

Homozygous or compound heterozygous mutations in fibulin-4 (FBLN4) lead to autosomal recessive cutis laxa type 1B (ARCL1B), a multisystem disorder characterized by significant cardiovascular abnormalities, including abnormal elastin assembly, arterial tortuosity, and aortic aneurysms. We sought to determine the consequences of a human disease-causing mutation in FBLN4 (E57K) on the cardiovascular system and vascular elastic fibers in a mouse model of ARCL1B. Fbln4E57K/E57K mice were hypertensive and developed arterial elongation, tortuosity, and ascending aortic aneurysms. Smooth muscle cell organization within the arterial wall of large conducting vessels was abnormal, and elastic fibers were fragmented and had a moth-eaten appearance. In contrast, vessel wall structure and elastic fiber integrity were normal in resistance/muscular arteries (renal, mesenteric, and saphenous). Elastin cross-linking and total elastin content were unchanged in large or small arteries, whereas elastic fiber architecture was abnormal in large vessels. While the E57K mutation did not affect Fbln4 mRNA levels, FBLN4 protein was lower in the ascending aorta of mutant animals compared to wild-type arteries but equivalent in mesenteric arteries. We found a differential role of FBLN4 in elastic fiber assembly, where it functions mainly in large conduit arteries. These results suggest that elastin assembly has different requirements depending on vessel type. Normal levels of elastin cross-links in mutant tissue call into question FBLN4's suggested role in mediating lysyl oxidase-elastin interactions. Future studies investigating tissue-specific elastic fiber assembly may lead to novel therapeutic interventions for ARCL1B and other disorders of elastic fiber assembly.


Assuntos
Artérias/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Aorta/metabolismo , Aneurisma Aórtico/metabolismo , Artérias/anormalidades , Sistema Cardiovascular/metabolismo , Cútis Laxa , Tecido Elástico/metabolismo , Elastina/metabolismo , Feminino , Humanos , Instabilidade Articular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Proteína-Lisina 6-Oxidase/metabolismo , Dermatopatias Genéticas/metabolismo , Malformações Vasculares/metabolismo
12.
Am J Physiol Heart Circ Physiol ; 309(5): H1008-16, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26232234

RESUMO

Increased arterial stiffness is a common characteristic of humans with Williams-Beuren syndrome and mouse models of elastin insufficiency. Arterial stiffness is associated with multiple negative cardiovascular outcomes, including myocardial infarction, stroke, and sudden death. Therefore, identifying therapeutic interventions that improve arterial stiffness in response to changes in elastin levels is of vital importance. The goal of this study was to determine the effect of chronic pharmacologic therapy with different classes of antihypertensive medications on arterial stiffness in elastin insufficiency. Elastin-insufficient mice 4-6 wk of age and wild-type littermates were subcutaneously implanted with osmotic micropumps delivering a continuous dose of one of the following: vehicle, losartan, nicardipine, or propranolol for 8 wk. At the end of treatment period, arterial blood pressure and large artery compliance and remodeling were assessed. Our results show that losartan and nicardipine treatment lowered blood pressure and pulse pressure in elastin-insufficient mice. Elastin and collagen content of abdominal aortas as well as ascending aorta and carotid artery biomechanics were not affected by any of the drug treatments in either genotype. By reducing pulse pressure and shifting the working pressure range of an artery to a more compliant region of the pressure-diameter curve, antihypertensive medications may mitigate the consequences of arterial stiffness, an effect that is drug class independent. These data emphasize the importance of early recognition and long-term management of hypertension in Williams-Beuren syndrome and elastin insufficiency.


Assuntos
Anti-Hipertensivos/uso terapêutico , Artérias/efeitos dos fármacos , Pressão Sanguínea , Rigidez Vascular/efeitos dos fármacos , Síndrome de Williams/tratamento farmacológico , Animais , Artérias/fisiopatologia , Elastina/deficiência , Elastina/genética , Losartan/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Nicardipino/uso terapêutico , Propranolol/uso terapêutico , Rigidez Vascular/genética , Síndrome de Williams/fisiopatologia
13.
J Biol Chem ; 288(40): 28869-80, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23963447

RESUMO

Microfibril-associated glycoprotein (MAGP) 1 and 2 are evolutionarily related but structurally divergent proteins that are components of microfibrils of the extracellular matrix. Using mice with a targeted inactivation of Mfap5, the gene for MAGP2 protein, we demonstrate that MAGPs have shared as well as unique functions in vivo. Mfap5(-/-) mice appear grossly normal, are fertile, and have no reduction in life span. Cardiopulmonary development is typical. The animals are normotensive and have vascular compliance comparable with age-matched wild-type mice, which is indicative of normal, functional elastic fibers. Loss of MAGP2 alone does not significantly alter bone mass or architecture, and loss of MAGP2 in tandem with loss of MAGP1 does not exacerbate MAGP1-dependent osteopenia. MAGP2-deficient mice are neutropenic, which contrasts with monocytopenia described in MAGP1-deficient animals. This suggests that MAGP1 and MAGP2 have discrete functions in hematopoiesis. In the cardiovascular system, MAGP1;MAGP2 double knockout mice (Mfap2(-/-);Mfap5(-/-)) show age-dependent aortic dilation. These findings indicate that MAGPs have shared primary functions in maintaining large vessel integrity. In solid phase binding assays, MAGP2 binds active TGFß1, TGFß2, and BMP2. Together, these data demonstrate that loss of MAGP2 expression in vivo has pleiotropic effects potentially related to the ability of MAGP2 to regulate growth factors or participate in cell signaling.


Assuntos
Proteínas Contráteis/deficiência , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/metabolismo , Pleiotropia Genética , Alelos , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Densidade Óssea , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Movimento Celular , Proteínas Contráteis/química , Éxons/genética , Proteínas da Matriz Extracelular/química , Marcação de Genes , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neutropenia/metabolismo , Neutropenia/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Tamanho do Órgão , Ligação Proteica , Fatores de Processamento de RNA , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
14.
J Cell Biochem ; 114(8): 1871-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23494777

RESUMO

The αvß3 integrin stimulates the resorptive capacity of the differentiated osteoclast (OC) by organizing its cytoskeleton via the tyrosine kinase, Syk. Thus, Syk-deficient OCs fails to spread or form actin rings, in vitro and in vivo. The Syk family of tyrosine kinases consists of Syk itself and Zap70 which are expressed by different cell types. Because of their structural similarity, and its compensatory properties in other cells, we asked if Zap70 can substitute for absence of Syk in OCs. While expression of Syk, as expected, normalizes the cytoskeletal abnormalities of Syk(-/-) OCs, Zap70 fails do so. In keeping with this observation, Syk, but not Zap70, rescues αvß3 integrin-induced SLP76 phosphorylation in Syk(-/-) OCs. Furthermore the kinase sequence of Syk partially rescues the Syk(-/-) phenotype but full normalization also requires its SH2 domains. Surprisingly, expression of Zap70 inhibits WT OC spreading, actin ring formation and bone resorptive activity, but not differentiation. In keeping with arrested cytoskeletal organization, Zap70 blocks integrin-activated endogenous Syk and Vav3, SLP76 phosphorylation. Such inhibition requires Zap70 kinase activity, as it is abolished by mutation of the Zap70 kinase domain. Thus, while the kinase domain of Syk is uniquely required for OC function that of Zap70 inhibits it.


Assuntos
Citoesqueleto/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteoclastos/enzimologia , Proteínas Tirosina Quinases/metabolismo , Proteína-Tirosina Quinase ZAP-70/metabolismo , Animais , Citoesqueleto/genética , Regulação da Expressão Gênica/fisiologia , Integrina alfa5/genética , Integrina alfa5/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Osteoclastos/citologia , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-vav/biossíntese , Quinase Syk , Proteína-Tirosina Quinase ZAP-70/genética
15.
J Biol Chem ; 288(8): 5268-77, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23300084

RESUMO

Type 1 phosphotidylinosotol-4 phosphate 5 kinase γ (PIP5KIγ) is central to generation of phosphotidylinosotol (4,5)P(2) (PI(4,5)P(2)). PIP5KIγ also participates in cytoskeletal organization by delivering talin to integrins, thereby enhancing their ligand binding capacity. As the cytoskeleton is pivotal to osteoclast function, we hypothesized that absence of PIP5KIγ would compromise their resorptive capacity. Absence of the kinase diminishes PI(4,5) abundance and desensitizes precursors to RANK ligand-stimulated differentiation. Thus, PIP5KIγ(-/-) osteoclasts are reduced in number in vitro and confirm physiological relevance in vivo. Despite reduced numbers, PIP5KIγ(-/-) osteoclasts surprisingly have normal cytoskeletons and effectively resorb bone. PIP5KIγ overexpression, which increases PI(4,5)P(2), also delays osteoclast differentiation and reduces cell number but in contrast to cells lacking the kinase, its excess disrupts the cytoskeleton. The cytoskeleton-disruptive effects of excess PIP5KIγ reflect its kinase activity and are independent of talin recognition. The combined arrested differentiation and disorganized cytoskeleton of PIP5KIγ-transduced osteoclasts compromises bone resorption. Thus, optimal PIP5KIγ and PI(4,5)P(2) expression, by osteoclasts, are essential for skeletal homeostasis.


Assuntos
Regulação Enzimológica da Expressão Gênica , Osteoclastos/citologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Animais , Transporte Biológico , Reabsorção Óssea , Cálcio/metabolismo , Diferenciação Celular , Ligantes , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoclastos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositóis/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Plasmídeos/metabolismo , Ligante RANK/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos
16.
J Bone Miner Res ; 27(12): 2490-500, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22807029

RESUMO

Osteoclastic bone resorption depends upon the cell's ability to organize its cytoskeleton via the αvß3 integrin and osteoclastogenic cytokines. Because paxillin associates with αvß3, we asked if it participates in skeletal degradation. Unlike deletion of other αvß3-associated cytoskeleton-regulating molecules, which impairs the cell's ability to spread, paxillin-deficient (Pax(-/-) ) osteoclasts, generated from embryonic stem cells, "superspread" in response to receptor activator of NF-κB ligand (RANKL) and form large, albeit dynamically atypical, actin bands. Despite their increased size, Pax(-/-) osteoclasts resorb bone poorly, excavating pits approximately one-third normal depth. Ligand-occupied αvß3 or RANKL promotes paxillin serine and tyrosine phosphorylation, the latter via cellular sarcoma (c-Src). The abnormal Pax(-/-) phenotype is rescued by wild-type (WT) paxillin but not that lacking its LD4 domain. In keeping with the appearance of mutant osteoclasts, WT paxillin, overexpressed in WT cells, contracts the cytoskeleton. Most importantly, the abnormal phenotype of Pax(-/-) osteoclasts likely represents failed RANKL-mediated delivery of myosin IIA to the actin cytoskeleton via the paxillin LD4 domain but is independent of tyrosine phosphorylation. Thus, in response to RANKL, paxillin associates with myosin IIA to contract the osteoclast cytoskeleton, thereby promoting its bone-degrading capacity.


Assuntos
Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Paxilina/farmacologia , Animais , Reabsorção Óssea/fisiopatologia , Humanos , Integrina alfaVbeta3/metabolismo , Camundongos , Miosina não Muscular Tipo IIA/metabolismo , Fosforilação , Ligante RANK/fisiologia
17.
J Mol Cell Cardiol ; 53(3): 437-45, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22796573

RESUMO

Transgenic mice overexpressing SUR1 and gain of function Kir6.2[∆N30, K185Q] K(ATP) channel subunits, under cardiac α-myosin heavy chain (αMHC) promoter control, demonstrate arrhythmia susceptibility and premature death. Pregnant mice, crossed to carry double transgenic progeny, which harbor high levels of both overexpressed subunits, exhibit the most extreme phenotype and do not deliver any double transgenic pups. To explore the fetal lethality and embryonic phenotype that result from K(ATP) overexpression, wild type (WT) and K(ATP) overexpressing embryonic cardiomyocytes were isolated, cultured and voltage-clamped using whole cell and excised patch clamp techniques. Whole mount embryonic imaging, Hematoxylin and Eosin (H&E) and α smooth muscle actin (αSMA) immunostaining were used to assess anatomy, histology and cardiac development in K(ATP) overexpressing and WT embryos. Double transgenic embryos developed in utero heart failure and 100% embryonic lethality by 11.5 days post conception (dpc). K(ATP) currents were detectable in both WT and K(ATP)-overexpressing embryonic cardiomyocytes, starting at early stages of cardiac development (9.5 dpc). In contrast to adult cardiomyocytes, WT and K(ATP)-overexpressing embryonic cardiomyocytes exhibit basal and spontaneous K(ATP) current, implying that these channels may be open and active under physiological conditions. At 9.5 dpc, live double transgenic embryos demonstrated normal looping pattern, although all cardiac structures were collapsed, probably representing failed, non-contractile chambers. In conclusion, K(ATP) channels are present and active in embryonic myocytes, and overexpression causes in utero heart failure and results in embryonic lethality. These results suggest that the K(ATP) channel may have an important physiological role during early cardiac development.


Assuntos
Perda do Embrião/genética , Genes Letais , Canais KATP/genética , Miocárdio/metabolismo , Animais , Feminino , Expressão Gênica , Coração/fisiopatologia , Átrios do Coração/enzimologia , Átrios do Coração/metabolismo , Átrios do Coração/fisiopatologia , Canais KATP/metabolismo , Camundongos , Camundongos Transgênicos , Gravidez
18.
J Biol Chem ; 287(26): 22055-67, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22573328

RESUMO

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.


Assuntos
Processamento Alternativo , Cútis Laxa/genética , Elastina/biossíntese , Elastina/genética , Mutação , Animais , Aorta/metabolismo , Cromossomos Artificiais Bacterianos , Reagentes de Ligações Cruzadas/química , Elasticidade , Elastina/metabolismo , Éxons , Fibroblastos/citologia , Mutação da Fase de Leitura , Genes Dominantes , Humanos , Camundongos , Camundongos Transgênicos , Estrutura Terciária de Proteína , RNA/química , Transgenes
19.
J Cell Biochem ; 113(1): 93-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898536

RESUMO

Microfibril-associated glycoprotein-1 (MAGP1), together with the fibrillins, are constitutive components of vertebrate microfibrils. Mice deficient in MAGP1 (murine MAGP1 knockout animals (Mfap2(-/-)); MAGP1Δ) is appropriate develop progressive osteopenia and reduced whole bone strength, and have elevated numbers of osteoclasts lining the bone surface. Our previous studies suggested that the increased osteoclast population was associated with elevated levels of receptor activator of NF-κB ligand (RANKL), a positive regulator of osteoclast differentiation. To explore the relationship between RANKL expression and osteoclast differentiation in MAGP1 deficiency, oophorectomy (OVX) was used to stimulate RANKL expression in both WT and MAGP1Δ animals. Bone loss following OVX was monitored using whole body DEXA and in vivo µCT. While WT mice exhibited significant bone loss following OVX, percent bone loss was reduced in MAGP1Δ mice. Further, serum RANKL levels rose significantly in OVX WT mice, whereas, there was only a modest increase in RANKL following OVX in the mutant mice due to already high baseline levels. Elevated RANKL expression was normalized when cultured MAGP1Δ osteoblasts were treated with a neutralizing antibody targeting free TGFß. These studies provide support for increased RANKL expression associated with MAGP1 deficiency and provide a link to altered TGF-ß signaling as a possible causative signaling pathway regulating RANKL expression in MAGP1Δ osteoblasts.


Assuntos
Reabsorção Óssea/metabolismo , Proteínas Contráteis/deficiência , Proteínas da Matriz Extracelular/deficiência , Ligante RANK/metabolismo , Animais , Doenças Ósseas Metabólicas/metabolismo , Reabsorção Óssea/genética , Diferenciação Celular , Células Cultivadas , Proteínas Contráteis/genética , Proteínas da Matriz Extracelular/genética , Feminino , Camundongos , Camundongos Knockout , Microfibrilas/fisiologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Ovariectomia , Fatores de Processamento de RNA , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
20.
J Biol Chem ; 286(52): 44926-36, 2011 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-22049077

RESUMO

Elastin haploinsufficiency causes the cardiovascular complications associated with Williams-Beuren syndrome and isolated supravalvular aortic stenosis. Significant variability exists in the vascular pathology in these individuals. Using the Eln(+/-) mouse, we sought to identify the source of this variability. Following outcrossing of C57Bl/6J Eln(+/-), two backgrounds were identified whose cardiovascular parameters deviated significantly from the parental strain. F1 progeny of the C57Bl/6J; Eln(+/-)x129X1/SvJ were more hypertensive and their arteries less compliant. In contrast, Eln(+/-) animals crossed to DBA/2J were protected from the pathologic changes associated with elastin insufficiency. Among the crosses, aortic elastin and collagen content did not correlate with quantitative vasculopathy traits. Quantitative trait locus analysis performed on F2 C57; Eln(+/-)x129 intercrosses identified highly significant peaks on chromosome 1 (LOD 9.7) for systolic blood pressure and on chromosome 9 (LOD 8.7) for aortic diameter. Additional peaks were identified that affect only Eln(+/-), including a region upstream of Eln on chromosome 5 (LOD 4.5). Bioinformatic analysis of the quantitative trait locus peaks revealed several interesting candidates, including Ren1, Ncf1, and Nos1; genes whose functions are unrelated to elastic fiber assembly, but whose effects may synergize with elastin insufficiency to predispose to hypertension and stiffer blood vessels. Real time RT-PCR studies show background-specific increased expression of Ncf1 (a subunit of the NOX2 NAPDH oxidase) that parallel the presence of increased oxidative stress in Eln(+/-) aortas. This finding raises the possibility that polymorphisms in genes affecting the generation of reactive oxygen species alter cardiovascular function in individuals with elastin haploinsufficiency through extrinsic noncomplementation.


Assuntos
Aorta/metabolismo , Elastina/metabolismo , Haploinsuficiência , Hipertensão/metabolismo , Síndrome de Williams/metabolismo , Animais , Aorta/patologia , Aorta/fisiopatologia , Pressão Sanguínea/genética , Cruzamentos Genéticos , Elastina/genética , Humanos , Hipertensão/genética , Hipertensão/patologia , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Mutantes , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Tamanho do Órgão , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Síndrome de Williams/genética , Síndrome de Williams/patologia , Síndrome de Williams/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...