Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood Adv ; 5(20): 3960-3974, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34500457

RESUMO

Bone marrow (BM) is the primary site of hematopoiesis and is responsible for a lifelong supply of all blood cell lineages. The process of hematopoiesis follows key intrinsic programs that also integrate instructive signals from the BM niche. First identified as an erythropoietin-potentiating factor, the tissue inhibitor of metalloproteinase (TIMP) protein family has expanded to 4 members and has widely come to be viewed as a classical regulator of tissue homeostasis. By virtue of metalloprotease inhibition, TIMPs not only regulate extracellular matrix turnover but also control growth factor bioavailability. The 4 mammalian TIMPs possess overlapping enzyme-inhibition profiles and have never been studied for their cumulative role in hematopoiesis. Here, we show that TIMPs are critical for postnatal B lymphopoiesis in the BM. TIMP-deficient mice have defective B-cell development arising at the pro-B-cell stage. Expression analysis of TIMPless hematopoietic cell subsets pointed to an altered B-cell program in the Lineage-Sca-1+c-Kit+ (LSK) cell fraction. Serial and competitive BM transplants identified a defect in TIMP-deficient hematopoietic stem and progenitor cells for B lymphopoiesis. In parallel, reverse BM transplants uncovered the extrinsic role of stromal TIMPs in pro- and pre-B-cell development. TIMP deficiency disrupted CXCL12 localization to LepR+ cells, and increased soluble CXCL12 within the BM niche. It also compromised the number and morphology of LepR+ cells. These data provide new evidence that TIMPs control the cellular and biochemical makeup of the BM niche and influence the LSK transcriptional program required for optimal B lymphopoiesis.


Assuntos
Células da Medula Óssea , Medula Óssea , Animais , Linfócitos B , Hematopoese , Camundongos , Inibidores Teciduais de Metaloproteinases/genética
2.
J Cell Biol ; 218(9): 3134-3152, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31371388

RESUMO

Regulated growth plate activity is essential for postnatal bone development and body stature, yet the systems regulating epiphyseal fusion are poorly understood. Here, we show that the tissue inhibitors of metalloprotease (TIMP) gene family is essential for normal bone growth after birth. Whole-body quadruple-knockout mice lacking all four TIMPs have growth plate closure in long bones, precipitating limb shortening, epiphyseal distortion, and widespread chondrodysplasia. We identify TIMP/FGF-2/IHH as a novel nexus underlying bone lengthening where TIMPs negatively regulate the release of FGF-2 from chondrocytes to allow IHH expression. Using a knock-in approach that combines MMP-resistant or ADAMTS-resistant aggrecans with TIMP deficiency, we uncouple growth plate activity in axial and appendicular bones. Thus, natural metalloprotease inhibitors are crucial regulators of chondrocyte maturation program, growth plate integrity, and skeletal proportionality. Furthermore, individual and combinatorial TIMP-deficient mice demonstrate the redundancy of metalloprotease inhibitor function in embryonic and postnatal development.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/metabolismo , Condrócitos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Lâmina de Crescimento/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Animais , Fator 2 de Crescimento de Fibroblastos/genética , Camundongos , Camundongos Knockout , Inibidores Teciduais de Metaloproteinases/genética
3.
J Bone Miner Res ; 34(1): 182-194, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30216540

RESUMO

Deregulated proteolysis invariably underlies most human diseases including bone pathologies. Metalloproteinases constitute the largest of the five protease families, and the metzincin metalloproteinases are inhibited by the four tissue inhibitors of metalloproteinase called TIMPs. We hypothesized that Timp genes are essential for skeletal homeostasis. We bred individual Timp knockout mice to generate unique mouse models, the quadruple Timp null strain (QT) as well as mice harboring only a single Timp3 allele (QT3+/- ). QT mice are grossly smaller and exhibit a dramatic reduction of trabeculae in long bones by µCT imaging with a corresponding increase in metalloproteinase activity. At the cellular level, Timp deficiency compromised differentiation markers, matrix deposition and mineralization in neonatal osteoblasts from calvariae, as well as the fibroblastic colony-forming unit (CFU-F) capacity of bone marrow-derived stromal cells. In contrast, we observed that osteoclasts were overactive in the Timp null state, consistent with the noted excessive bone resorption of QT bones. Immunohistochemistry (IHC) and immunofluorescence (IF) analyses of bone sections revealed higher Cathepsin K and RANKL signals upon Timp loss. Seeking the molecular mechanism, we identified abnormal TNFα bioactivity to be a central event in Timp-deficient mice. Specifically, TNFα triggered induction of the Wnt signaling inhibitor Dkk1 in the osteoblasts at the mRNA and protein levels, with a simultaneous increase in RANKL. Neutralizing TNFα antibody was capable of rescuing the induction of Dkk1 as well as RANKL. Therefore, the generation of novel Timp-deficient systems allowed us to uncover the essential and collective function of TIMP proteins in mammalian long-bone homeostasis. Moreover, our study discovers a functional TIMP/metalloproteinase-TNFα-Dkk1/RANKL nexus for optimal control of the bone microenvironment, which dictates coexistence of the osteoblast and osteoclast lineages. © 2018 American Society for Bone and Mineral Research.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteases/metabolismo , Crânio/metabolismo , Inibidores Teciduais de Metaloproteinases/deficiência , Fator de Necrose Tumoral alfa/metabolismo , Via de Sinalização Wnt , Animais , Catepsina K/genética , Catepsina K/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Metaloproteases/genética , Camundongos , Camundongos Knockout , Tamanho do Órgão , Ligante RANK/genética , Ligante RANK/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Fator de Necrose Tumoral alfa/genética
4.
Nat Cell Biol ; 16(9): 889-901, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25150980

RESUMO

Cancer-associated fibroblasts (CAFs) drive tumour progression, but the emergence of this cell state is poorly understood. A broad spectrum of metalloproteinases, controlled by the Timp gene family, influence the tumour microenvironment in human cancers. Here, we generate quadruple TIMP knockout (TIMPless) fibroblasts to unleash metalloproteinase activity within the tumour-stromal compartment and show that complete Timp loss is sufficient for the acquisition of hallmark CAF functions. Exosomes produced by TIMPless fibroblasts induce cancer cell motility and cancer stem cell markers. The proteome of these exosomes is enriched in extracellular matrix proteins and the metalloproteinase ADAM10. Exosomal ADAM10 increases aldehyde dehydrogenase expression in breast cancer cells through Notch receptor activation and enhances motility through the GTPase RhoA. Moreover, ADAM10 knockdown in TIMPless fibroblasts abrogates their CAF function. Importantly, human CAFs secrete ADAM10-rich exosomes that promote cell motility and activate RhoA and Notch signalling in cancer cells. Thus, Timps suppress cancer stroma where activated-fibroblast-secreted exosomes impact tumour progression.


Assuntos
Fibroblastos/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Inibidores Teciduais de Metaloproteinases/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Exossomos/fisiologia , Feminino , Fibroblastos/patologia , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Mamárias Experimentais/enzimologia , Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Fenótipo , Transdução de Sinais , Inibidores Teciduais de Metaloproteinases/deficiência , Carga Tumoral
5.
Biochim Biophys Acta ; 1803(1): 121-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19616584

RESUMO

The metalloproteinase family includes MMP, ADAM and ADAMTS proteases. Mice deficient in individual or pairs of metalloproteinases have been generated, and a number of these genetic models spontaneously develop skeletal abnormalities. Here we review metalloproteinase function in endochondral and intramembranous ossification, as well as in postnatal bone remodeling. We highlight how metalloproteinases enable interactions between distinct bone cell types and how this communication contributes to the skeletal phenotypes observed in knockout mice. In addition to the physiological actions of metalloproteinases in the skeletal system, the experimental manipulation of metalloproteinase-deficient mice has revealed substantial roles for these enzymes in osteoarthritis and rheumatoid arthritis. MMP, ADAM and ADAMTS proteases thus emerge as key players in the development and homeostasis of the skeletal system.


Assuntos
Doenças Ósseas/enzimologia , Osso e Ossos/enzimologia , Osso e Ossos/patologia , Metaloproteases/metabolismo , Animais , Doenças Ósseas/fisiopatologia , Remodelação Óssea , Comunicação Celular , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...