Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 221(6)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38597952

RESUMO

Epithelium-derived cytokines or alarmins, such as interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP), are major players in type 2 immunity and asthma. Here, we demonstrate that TNF-like ligand 1A (TL1A) is an epithelial alarmin, constitutively expressed in alveolar epithelium at steady state in both mice and humans, which cooperates with IL-33 for early induction of IL-9high ILC2s during the initiation of allergic airway inflammation. Upon synergistic activation by IL-33 and TL1A, lung ILC2s acquire a transient IL-9highGATA3low "ILC9" phenotype and produce prodigious amounts of IL-9. A combination of large-scale proteomic analyses, lung intravital microscopy, and adoptive transfer of ILC9 cells revealed that high IL-9 expression distinguishes a multicytokine-producing state-of-activated ILC2s with an increased capacity to initiate IL-5-dependent allergic airway inflammation. Similar to IL-33 and TSLP, TL1A is expressed in airway basal cells in healthy and asthmatic human lungs. Together, these results indicate that TL1A is an epithelium-derived cytokine and an important cofactor of IL-33 in the airways.


Assuntos
Asma , Interleucina-33 , Animais , Humanos , Camundongos , Alarminas , Citocinas , Imunidade Inata , Inflamação , Interleucina-9 , Linfócitos , Proteômica
2.
STAR Protoc ; 3(2): 101444, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35677615

RESUMO

Here, we present a protocol for flow cytometry analysis of endothelial cells (ECs) and CD8+ T cells in murine tumor models, at baseline and after cancer immunotherapy with anti-PD-1/anti-CTLA-4 antibodies. We provide gating strategies for identification of specific cell subsets including ECs from tumor-associated high endothelial venules (TA-HEVs), stem-like, and terminally exhausted CD8+ T cells. This protocol represents a valuable tool for the analysis of rare subsets of tumor ECs and CD8+ T cells with critical roles in antitumor immunity. For complete details on the use and execution of this protocol, please refer to Asrir et al. (2022).


Assuntos
Neoplasias , Receptor de Morte Celular Programada 1 , Animais , Linfócitos T CD8-Positivos , Células Endoteliais , Citometria de Fluxo , Imunoterapia/métodos , Camundongos , Neoplasias/terapia
3.
Cancer Cell ; 40(3): 318-334.e9, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35120598

RESUMO

Recruitment of lymphocytes into tumors is critical for anti-tumor immunity and efficacious immunotherapy. We show in murine models that tumor-associated high endothelial venules (TA-HEVs) are major sites of lymphocyte entry into tumors at baseline and upon treatment with anti-PD-1/anti-CTLA-4 immune checkpoint blockade (ICB). TA-HEV endothelial cells (TA-HECs) derive from post-capillary venules, co-express MECA-79+ HEV sialomucins and E/P-selectins, and are associated with homing and infiltration into tumors of various T cell subsets. Intravital microscopy further shows that TA-HEVs are the main sites of lymphocyte arrest and extravasation into ICB-treated tumors. Increasing TA-HEC frequency and maturation increases the proportion of tumor-infiltrating stem-like CD8+ T cells, and ameliorates ICB efficacy. Analysis of tumor biopsies from 93 patients with metastatic melanoma reveals that TA-HEVs are predictive of better response and survival upon treatment with anti-PD-1/anti-CTLA-4 combination. These studies provide critical insights into the mechanisms governing lymphocyte trafficking in cancer immunity and immunotherapy.


Assuntos
Melanoma , Receptor de Morte Celular Programada 1 , Animais , Linfócitos T CD8-Positivos , Antígeno CTLA-4 , Células Endoteliais , Humanos , Fatores Imunológicos , Imunoterapia , Linfócitos do Interstício Tumoral , Melanoma/patologia , Camundongos , Subpopulações de Linfócitos T , Vênulas/patologia
4.
FASEB J ; 27(9): 3608-18, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23742809

RESUMO

In osteoclasts, Src controls podosome organization and bone degradation, which leads to an osteopetrotic phenotype in src(-/-) mice. Since this phenotype was even more severe in src(-/-)hck(-/-) mice, we examined the individual contribution of Hck in bone homeostasis. Compared to wt mice, hck(-/-) mice exhibited an osteopetrotic phenotype characterized by an increased density of trabecular bone and decreased bone degradation, although osteoclastogenesis was not impaired. Podosome organization and matrix degradation were found to be defective in hck(-/-) osteoclast precursors (preosteoclast) but were normal in mature hck(-/-) osteoclasts, probably through compensation by Src, which was specifically overexpressed in mature osteoclasts. As a consequence of podosome defects, the 3-dimensional migration of hck(-/-) preosteoclasts was strongly affected in vitro. In vivo, this translated by altered bone homing of preosteoclasts in hck(-/-) mice: in metatarsals of 1-wk-old mice, when bone formation strongly depends on the recruitment of these cells, reduced numbers of osteoclasts and abnormal developing trabecular bone were observed. This phenotype was still detectable in adults. In summmary, Hck is one of the very few effectors of preosteoclast recruitment described to date and thereby plays a critical role in bone remodeling.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Movimento Celular/fisiologia , Osteoclastos/citologia , Osteopetrose/metabolismo , Proteínas Proto-Oncogênicas c-hck/metabolismo , Animais , Movimento Celular/genética , Células Cultivadas , Feminino , Homeostase/genética , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/metabolismo , Osteopetrose/genética , Proteínas Proto-Oncogênicas c-hck/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
5.
Gut ; 62(12): 1714-23, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23172891

RESUMO

OBJECTIVE: Inflammatory bowel diseases (IBD) have been intrinsically linked to a deregulated cytokine network, but novel therapeutic principles are urgently needed. Here we identify the interleukin (IL)-33 and its receptor ST2 as key negative regulators of wound healing and permeability in the colon of mice. DESIGN: Expression of IL-33 and ST2 was determined by qRT-PCR, ELISA, immunohistochemistry and western-blot analysis. Wild-type and St2(-/-) mice were used in wound healing experiments and in two experimental models of IBD triggered by 2,4,6-trinitrobenzene sulphonic acid or dextran sodium sulphate (DSS). Neutralisation of ST2 was performed by using a specific blocking antibody. RESULTS: Nuclear localisation and enhanced expression of IL-33 in myofibroblasts and enterocytes was linked to disease involvement independently of inflammation, while the expression of ST2 was primarily restricted to the colonic epithelia. In two experimental models of IBD, genetic ablation of ST2 significantly improved signs of colitis, while a sustained epithelial expression of the cyto-protective factor connexin-43 was observed in DSS-treated St2-deficient mice. Unexpectedly, absence of ST2 in non-hematopoietic cells was sufficient to protect against colitis. Consistently, specific inhibition of endogenous ST2-mediated signalling by treatment with neutralising antibody improved DSS-induced colitis. In addition, IL-33 treatment impaired epithelial barrier permeability in vitro and in vivo, whereas absence of ST2 enhanced wound healing response upon acute mechanical injury in the colon. CONCLUSIONS: Our study unveiled a novel non-hematopoietic function of IL-33 in epithelial barrier function and wound healing. Therefore, blocking the IL-33/ST2 axis may represent an efficient therapy in IBD.


Assuntos
Colite Ulcerativa/etiologia , Interleucinas/fisiologia , Receptores de Interleucina/fisiologia , Animais , Western Blotting , Células CACO-2 , Colite Ulcerativa/fisiopatologia , Colite Ulcerativa/terapia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Reação em Cadeia da Polimerase em Tempo Real , Cicatrização/fisiologia
6.
J Immunol ; 188(7): 3488-95, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22371395

RESUMO

IL-33 (previously known as NF from high endothelial venules) is an IL-1 family cytokine that signals through the ST2 receptor and drives cytokine production in mast cells, basophils, eosinophils, invariant NKT and NK cells, Th2 lymphocytes, and type 2 innate immune cells (natural helper cells, nuocytes, and innate helper 2 cells). Little is known about endogenous IL-33; for instance, the cellular sources of IL-33 in mouse tissues have not yet been defined. In this study, we generated an Il-33-LacZ gene trap reporter strain (Il-33(Gt/Gt)) and used this novel tool to analyze expression of endogenous IL-33 in vivo. We found that the Il-33 promoter exhibits constitutive activity in mouse lymphoid organs, epithelial barrier tissues, brain, and embryos. Immunostaining with anti-IL-33 Abs, using Il-33(Gt/Gt) (Il-33-deficient) mice as control, revealed that endogenous IL-33 protein is highly expressed in mouse epithelial barrier tissues, including stratified squamous epithelia from vagina and skin, as well as cuboidal epithelium from lung, stomach, and salivary gland. Constitutive expression of IL-33 was not detected in blood vessels, revealing the existence of species-specific differences between humans and mice. Importantly, IL-33 protein was always localized in the nucleus of producing cells with no evidence for cytoplasmic localization. Finally, strong expression of the Il-33-LacZ reporter was also observed in inflamed tissues, in the liver during LPS-induced endotoxin shock, and in the lung alveoli during papain-induced allergic airway inflammation. Together, our findings support the possibility that IL-33 may function as a nuclear alarmin to alert the innate immune system after injury or infection in epithelial barrier tissues.


Assuntos
Encéfalo/metabolismo , Núcleo Celular/metabolismo , Células Epiteliais/metabolismo , Inflamação/metabolismo , Interleucinas/fisiologia , Tecido Linfoide/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Proteínas Fetais/biossíntese , Proteínas Fetais/genética , Genes Reporter , Humanos , Interleucina-33 , Interleucinas/biossíntese , Interleucinas/genética , Óperon Lac , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Regiões Promotoras Genéticas/genética , Eosinofilia Pulmonar/induzido quimicamente , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/metabolismo , Choque Séptico/imunologia , Choque Séptico/metabolismo , Especificidade da Espécie
7.
J Biol Chem ; 285(18): 13364-71, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20200153

RESUMO

THAP1 is a sequence-specific DNA binding factor that regulates cell proliferation through modulation of target genes such as the cell cycle-specific gene RRM1. Mutations in the THAP1 DNA binding domain, an atypical zinc finger (THAP-zf), have recently been found to cause DYT6 dystonia, a neurological disease characterized by twisting movements and abnormal postures. In this study, we report that THAP1 shares sequence characteristics, in vivo expression patterns and protein partners with THAP3, another THAP-zf protein. Proteomic analyses identified HCF-1, a potent transcriptional coactivator and cell cycle regulator, and O-GlcNAc transferase (OGT), the enzyme that catalyzes the addition of O-GlcNAc, as major cellular partners of THAP3. THAP3 interacts with HCF-1 through a consensus HCF-1-binding motif (HBM), a motif that is also present in THAP1. Accordingly, THAP1 was found to bind HCF-1 in vitro and to associate with HCF-1 and OGT in vivo. THAP1 and THAP3 belong to a large family of HCF-1 binding factors since seven other members of the human THAP-zf protein family were identified, which harbor evolutionary conserved HBMs and bind to HCF-1. Chromatin immunoprecipitation (ChIP) assays and RNA interference experiments showed that endogenous THAP1 mediates the recruitment of HCF-1 to the RRM1 promoter during endothelial cell proliferation and that HCF-1 is essential for transcriptional activation of RRM1. Together, our findings suggest HCF-1 is an important cofactor for THAP1. Interestingly, our results also provide an unexpected link between DYT6 and DYT3 (X-linked dystonia-parkinsonism) dystonias because the gene encoding the THAP1/DYT6 protein partner OGT maps within the DYT3 critical region on Xq13.1.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Cromossomos Humanos X/metabolismo , Proteínas de Ligação a DNA/metabolismo , Distonia/metabolismo , Fator C1 de Célula Hospedeira/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Acetilglucosamina , Motivos de Aminoácidos , Proteínas Reguladoras de Apoptose/genética , Proliferação de Células , Cromossomos Humanos X/genética , Proteínas de Ligação a DNA/genética , Distonia/genética , Células Endoteliais , Doenças Genéticas Ligadas ao Cromossomo X , Células HeLa , Fator C1 de Célula Hospedeira/genética , Humanos , N-Acetilglucosaminiltransferases/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteômica , Ribonucleosídeo Difosfato Redutase , Transcrição Gênica/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Dedos de Zinco
8.
Dis Model Mech ; 3(3-4): 224-35, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20142327

RESUMO

Long bone development depends on endochondral bone formation, a complex process requiring exquisite balance between hypertrophic cartilage (HC) formation and its ossification. Dysregulation of this process may result in skeletal dysplasias and heterotopic ossification. Endochondral ossification requires the precise orchestration of HC vascularization, extracellular matrix remodeling, and the recruitment of osteoclasts and osteoblasts. Matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) and osteoclasts have all been shown to regulate endochondral ossification, but how their function interrelates is not known. We have investigated the functional relationship among these regulators of endochondral ossification, demonstrating that they have complementary but non-overlapping functions. MMP-9, VEGF and osteoclast deficiency all cause impaired growth plate ossification resulting in the accumulation of HC. VEGF mRNA and protein expression are increased at the MMP-9-/- growth plate, and VEGF activity contributes to endochondral ossification since sequestration of VEGF by soluble receptors results in further inhibition of growth plate vascularization and ossification. However, VEGF bioavailability is still limited in MMP-9 deficiency, as exogenous VEGF is able to rescue the MMP-9-/- phenotype, demonstrating that MMP-9 may partially, but not fully, regulate VEGF bioavailability. The organization of the HC extracellular matrix at the MMP-9-/- growth plate is altered, supporting a role for MMP-9 in HC remodeling. Inhibition of VEGF impairs osteoclast recruitment, whereas MMP-9 deficiency leads to an accumulation of osteoclasts at the chondro-osseous junction. Growth plate ossification in osteoclast-deficient mice is impaired in the presence of normal MMP-9 expression, indicating that other osteoclastic functions are also necessary. Our data delineate the complementary interplay between MMP-9, VEGF and osteoclast function that is necessary for normal endochondral bone formation and provide a molecular framework for investigating the molecular defects contributing to disorders of endochondral bone formation.


Assuntos
Condrócitos/patologia , Metaloproteinase 9 da Matriz/metabolismo , Osteoclastos/enzimologia , Osteoclastos/patologia , Osteogênese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fosfatase Ácida/metabolismo , Animais , Animais Recém-Nascidos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/enzimologia , Osso e Ossos/patologia , Osso e Ossos/ultraestrutura , Cartilagem/patologia , Cartilagem/ultraestrutura , Condrócitos/efeitos dos fármacos , Condrócitos/enzimologia , Condrócitos/ultraestrutura , Colágeno Tipo II/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/enzimologia , Lâmina de Crescimento/patologia , Lâmina de Crescimento/ultraestrutura , Humanos , Hipertrofia , Isoenzimas/metabolismo , Metaloproteinase 9 da Matriz/deficiência , Inibidores de Metaloproteinases de Matriz , Camundongos , Modelos Biológicos , Osteoclastos/efeitos dos fármacos , Osteoclastos/ultraestrutura , Osteogênese/efeitos dos fármacos , Fenótipo , Inibidores de Proteases/farmacologia , Fosfatase Ácida Resistente a Tartarato , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/farmacologia
9.
PLoS One ; 3(10): e3331, 2008 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-18836528

RESUMO

BACKGROUND: Interleukin-33 (IL-33) is an IL-1-like cytokine ligand for the IL-1 receptor-related protein ST2, that activates mast cells and Th2 lymphocytes, and induces production of Th2-associated cytokines in vivo. We initially discovered IL-33 as a nuclear factor (NF-HEV) abundantly expressed in high endothelial venules from lymphoid organs, that associates with chromatin and exhibits transcriptional regulatory properties. This suggested that, similarly to IL-1alpha and chromatin-associated cytokine HMGB1, IL-33 may act as both a cytokine and a nuclear factor. Although the activity of recombinant IL-33 has been well characterized, little is known yet about the expression pattern of endogenous IL-33 in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that IL-33 is constitutively and abundantly expressed in normal human tissues. Using a combination of human tissue microarrays and IL-33 monoclonal and polyclonal antibodies, we found that IL-33 is a novel nuclear marker of the endothelium widely expressed along the vascular tree. We observed abundant nuclear expression of IL-33 in endothelial cells from both large and small blood vessels in most normal human tissues, as well as in human tumors. In addition to endothelium, we also found constitutive nuclear expression of IL-33 in fibroblastic reticular cells of lymphoid tissues, and epithelial cells of tissues exposed to the environment, including skin keratinocytes and epithelial cells of the stomach, tonsillar crypts and salivary glands. CONCLUSIONS/SIGNIFICANCE: Together, our results indicate that, unlike inducible cytokines, IL-33 is constitutively expressed in normal human tissues. In addition, they reveal that endothelial cells and epithelial cells constitute major sources of IL-33 in vivo. Based on these findings, we speculate that IL-33 may function, similarly to the prototype 'alarmin' HMGB1, as an endogenous 'danger' signal to alert the immune system after endothelial or epithelial cell damage during trauma or infection.


Assuntos
Núcleo Celular/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-1/metabolismo , Interleucinas/metabolismo , Actinas/metabolismo , Citocinas/genética , Desmina/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Corantes Fluorescentes/metabolismo , Humanos , Imuno-Histoquímica , Indóis/metabolismo , Interleucina-1/genética , Interleucina-33 , Interleucinas/genética
10.
Mol Cell Proteomics ; 6(9): 1621-37, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17533220

RESUMO

Proteomics strategies based on nanoflow (nano-) LC-MS/MS allow the identification of hundreds to thousands of proteins in complex mixtures. When combined with protein isotopic labeling, quantitative comparison of the proteome from different samples can be achieved using these approaches. However, bioinformatics analysis of the data remains a bottleneck in large scale quantitative proteomics studies. Here we present a new software named Mascot File Parsing and Quantification (MFPaQ) that easily processes the results of the Mascot search engine and performs protein quantification in the case of isotopic labeling experiments using either the ICAT or SILAC (stable isotope labeling with amino acids in cell culture) method. This new tool provides a convenient interface to retrieve Mascot protein lists; sort them according to Mascot scoring or to user-defined criteria based on the number, the score, and the rank of identified peptides; and to validate the results. Moreover the software extracts quantitative data from raw files obtained by nano-LC-MS/MS, calculates peptide ratios, and generates a non-redundant list of proteins identified in a multisearch experiment with their calculated averaged and normalized ratio. Here we apply this software to the proteomics analysis of membrane proteins from primary human endothelial cells (ECs), a cell type involved in many physiological and pathological processes including chronic inflammatory diseases such as rheumatoid arthritis. We analyzed the EC membrane proteome and set up methods for quantitative analysis of this proteome by ICAT labeling. EC microsomal proteins were fractionated and analyzed by nano-LC-MS/MS, and database searches were performed with Mascot. Data validation and clustering of proteins were performed with MFPaQ, which allowed identification of more than 600 unique proteins. The software was also successfully used in a quantitative differential proteomics analysis of the EC membrane proteome after stimulation with a combination of proinflammatory mediators (tumor necrosis factor-alpha, interferon-gamma, and lymphotoxin alpha/beta) that resulted in the identification of a full spectrum of EC membrane proteins regulated by inflammation.


Assuntos
Membrana Celular/metabolismo , Células Endoteliais/metabolismo , Espectrometria de Massas/métodos , Proteômica/instrumentação , Proteômica/métodos , Cromatografia Líquida/métodos , Biologia Computacional , Citocinas/metabolismo , Endotélio Vascular/citologia , Humanos , Inflamação , Internet , Microssomos/metabolismo , Peptídeos/química , Software , Veias Umbilicais/citologia
11.
Proc Natl Acad Sci U S A ; 104(1): 282-7, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17185418

RESUMO

Recent studies indicate that IL-1alpha functions intracellularly in pathways independent of its cell surface receptors by translocating to the nucleus and regulating transcription. Similarly, the chromatin-associated protein HMGB1 acts as both a nuclear factor and a secreted proinflammatory cytokine. Here, we show that IL-33, an IL-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines, is an endothelium-derived, chromatin-associated nuclear factor with transcriptional repressor properties. We found that IL-33 is identical to NF-HEV, a nuclear factor preferentially expressed in high endothelial venules (HEV), that we previously characterized. Accordingly, in situ hybridization demonstrated that endothelial cells constitute a major source of IL-33 mRNA in chronically inflamed tissues from patients with rheumatoid arthritis and Crohn's disease. Immunostaining with three distinct antisera, directed against the N-terminal part and IL-1-like C-terminal domain, revealed that IL-33 is a heterochromatin-associated nuclear factor in HEV endothelial cells in vivo. Association of IL-33 with heterochromatin was also observed in human and mouse cells under living conditions. In addition, colocalization of IL-33 with mitotic chromatin was noted. Nuclear localization, heterochromatin-association, and targeting to mitotic chromosomes were all found to be mediated by an evolutionarily conserved homeodomain-like helix-turn-helix motif within the IL-33 N-terminal part. Finally, IL-33 was found to possess transcriptional repressor properties, associated with the homeodomain-like helix-turn-helix motif. Together, these data suggest that, similarly to IL1alpha and HMGB1, IL-33 is a dual function protein that may function as both a proinflammatory cytokine and an intracellular nuclear factor with transcriptional regulatory properties.


Assuntos
Heterocromatina/química , Interleucinas/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Nucleares/fisiologia , Receptores de Superfície Celular/fisiologia , Células 3T3 , Motivos de Aminoácidos , Animais , Artrite Reumatoide/metabolismo , Sequência Conservada , Doença de Crohn/metabolismo , Células Endoteliais/metabolismo , Células HeLa , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/química , Interleucinas/genética , Camundongos , Mitose , Sinais de Localização Nuclear , RNA Mensageiro/análise , Receptores de Interleucina , Proteínas Repressoras/fisiologia
12.
Mol Biol Cell ; 16(6): 3028-39, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15800063

RESUMO

Endochondral bone formation is characterized by the progressive replacement of a cartilage anlagen by bone at the growth plate with a tight balance between the rates of chondrocyte proliferation, differentiation, and cell death. Deficiency of matrix metalloproteinase-9 (MMP-9) leads to an accumulation of late hypertrophic chondrocytes. We found that galectin-3, an in vitro substrate of MMP-9, accumulates in the late hypertrophic chondrocytes and their surrounding extracellular matrix in the expanded hypertrophic cartilage zone. Treatment of wild-type embryonic metatarsals in culture with full-length galectin-3, but not galectin-3 cleaved by MMP-9, mimicked the embryonic phenotype of Mmp-9 null mice, with an increased hypertrophic zone and decreased osteoclast recruitment. These results indicate that extracellular galectin-3 could be an endogenous substrate of MMP-9 that acts downstream to regulate hypertrophic chondrocyte death and osteoclast recruitment during endochondral bone formation. Thus, the disruption of growth plate homeostasis in Mmp-9 null mice links galectin-3 and MMP-9 in the regulation of the clearance of late chondrocytes through regulation of their terminal differentiation.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/enzimologia , Galectina 3/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Animais , Western Blotting , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Diferenciação Celular , Células Cultivadas , Condrócitos/metabolismo , Matriz Extracelular/metabolismo , Lâmina de Crescimento/citologia , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/crescimento & desenvolvimento , Lâmina de Crescimento/metabolismo , Úmero/citologia , Úmero/embriologia , Imuno-Histoquímica , Hibridização In Situ , Ossos do Metatarso/citologia , Ossos do Metatarso/embriologia , Camundongos , Camundongos Mutantes , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo
13.
Development ; 131(23): 5883-95, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15539485

RESUMO

The assembly and degradation of extracellular matrix (ECM) molecules are crucial processes during bone development. In this study, we show that ECM remodeling is a critical rate-limiting step in endochondral bone formation. Matrix metalloproteinase (MMP) 13 (collagenase 3) is poised to play a crucial role in bone formation and remodeling because of its expression both in terminal hypertrophic chondrocytes in the growth plate and in osteoblasts. Moreover, a mutation in the human MMP13 gene causes the Missouri variant of spondyloepimetaphyseal dysplasia. Inactivation of Mmp13 in mice through homologous recombination led to abnormal skeletal growth plate development. Chondrocytes differentiated normally but their exit from the growth plate was delayed. The severity of the Mmp13- null growth plate phenotype increased until about 5 weeks and completely resolved by 12 weeks of age. Mmp13-null mice had increased trabecular bone, which persisted for months. Conditional inactivation of Mmp13 in chondrocytes and osteoblasts showed that increases in trabecular bone occur independently of the improper cartilage ECM degradation caused by Mmp13 deficiency in late hypertrophic chondrocytes. Our studies identified the two major components of the cartilage ECM, collagen type II and aggrecan, as in vivo substrates for MMP13. We found that degradation of cartilage collagen and aggrecan is a coordinated process in which MMP13 works synergistically with MMP9. Mice lacking both MMP13 and MMP9 had severely impaired endochondral bone, characterized by diminished ECM remodeling, prolonged chondrocyte survival, delayed vascular recruitment and defective trabecular bone formation (resulting in drastically shortened bones). These data support the hypothesis that proper ECM remodeling is the dominant rate-limiting process for programmed cell death, angiogenesis and osteoblast recruitment during normal skeletal morphogenesis.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/enzimologia , Colagenases/genética , Colagenases/fisiologia , Animais , Osso e Ossos/anormalidades , Osso e Ossos/metabolismo , Bromodesoxiuridina/farmacologia , Cartilagem/metabolismo , Diferenciação Celular , Células Cultivadas , Condrócitos/metabolismo , Colagenases/metabolismo , Matriz Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Metaloproteinase 13 da Matriz , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Modelos Genéticos , Mutação , Neovascularização Patológica , Fenótipo , Recombinação Genética , Fatores de Tempo , Tomografia Computadorizada por Raios X , Transgenes
14.
Trends Cell Biol ; 14(2): 86-93, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15102440

RESUMO

Endochondral ossification, the process by which most of the skeleton is formed, is a powerful system for studying various aspects of the biological response to degraded extracellular matrix (ECM). In addition, the dependence of endochondral ossification upon neovascularization and continuous ECM remodeling provides a good model for studying the role of the matrix metalloproteases (MMPs) not only as simple effectors of ECM degradation but also as regulators of active signal-inducers for the initiation of endochondral ossification. The daunting task of elucidating their specific role during endochondral ossification has been facilitated by the development of mice deficient for various members of this family. Here, we discuss the ECM and its remodeling as one level of molecular regulation for the process of endochondral ossification, with special attention to the MMPs.


Assuntos
Osso e Ossos/metabolismo , Matriz Extracelular/metabolismo , Osteogênese/fisiologia , Animais , Osso e Ossos/irrigação sanguínea , Osso e Ossos/citologia , Cartilagem/citologia , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Diferenciação Celular/fisiologia , Condrócitos/citologia , Condrócitos/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia
15.
Ann N Y Acad Sci ; 995: 109-16, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12814943

RESUMO

Matrix metalloproteinases (MMPs) degrade most components of the extracellular matrix (ECM), as well as many non-ECM molecules. MMPs participate in (1). degradation of ECM to allow cell migration; (2). alteration of the ECM microenvironment resulting in alteration in cellular behavior; (3). modulation of biologically active molecules by direct cleavage or release from ECM stores; (4). regulation of the activity of other proteases; and (5). cell attachment, proliferation, differentiation, and apoptosis. We have sought to understand the role of MMPs during development and tissue repair in transgenic mice. Endochondral bone formation presents a particularly interesting developmental challenge. During this process, an avascular tissue (cartilage) is converted into one of the most highly vascularized tissues (bone) in the vertebrate body. Ossification begins with invasion of the calcified hypertrophic cartilage by capillaries. Apoptosis of the terminal hypertrophic chondrocytes, degradation of the cartilage matrix, and deposition of bone matrix by osteoblasts accompanies neovascularization of the growth plate. Remodeling of ECM results in a cavity filled with vascular channels containing hematopoietic cells. Our results reveal that MMP9, MMP13, and vascular endothelial growth factor are key regulators for the remodeling of the skeletal tissues. They coordinate not only matrix degradation, but also the recruitment and differentiation of endothelial cells, osteoclasts, chondroclasts, and osteoprogenitors.


Assuntos
Osso e Ossos/embriologia , Metaloproteinases da Matriz/fisiologia , Osteogênese , Animais , Medula Óssea/enzimologia , Osso e Ossos/anatomia & histologia , Osso e Ossos/fisiologia , Hematopoese , Metaloproteinase 9 da Matriz/genética , Metaloproteinases da Matriz/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfogênese , Cicatrização
16.
Ann N Y Acad Sci ; 995: 140-50, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12814946

RESUMO

The cartilage extracellular matrix is composed of a dense collagen network that entraps a range of other specialized proteins important for the proper formation and function of the tissue. Loss of two abundant cartilage components, type II collagen and perlecan, has drastic effects on skeletal development. Both collagen II and perlecan mutants have severe and lethal chondrodysplasia characterized by disorganized growth plate, lack of collagen network, defective endochondral bone formation, and abnormal intervertebral disk development. To test whether the reduced collagen density in the perlecan-null cartilage is due to enhanced activity of collagen-degrading proteinases, we have analyzed gelatinase expression and activity in the mutant tissue. Immunohistochemical analysis revealed a weak, but clear, expansion of MMP-9 deposition into the hypertrophic zone of the perlecan-null growth plate. However, in situ and SDS-PAGE zymography showed that the activity of gelatinases (MMP-2 and MMP-9) is not altered in perlecan-null cartilage, suggesting that they are not primarily linked to the reduced fibrillar network observed in the mutant. Likewise, intercrossing of perlecan mutants onto an MMP-9-null background could not rescue the ultrastructural abnormalities of the perlecan-deficient cartilage.


Assuntos
Osso e Ossos/embriologia , Colágeno Tipo II/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Osteogênese , Animais , Osso e Ossos/anatomia & histologia , Cartilagem/embriologia , Cartilagem/ultraestrutura , Colágeno Tipo II/genética , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/ultraestrutura , Matriz Extracelular/ultraestrutura , Colágenos Fibrilares/ultraestrutura , Gelatinases/metabolismo , Proteoglicanas de Heparan Sulfato/genética , Camundongos , Camundongos Knockout , Osteocondrodisplasias/etiologia
17.
FASEB J ; 17(11): 1520-2, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12709411

RESUMO

Vascular endothelial growth factor (VEGF), a major factor mediating endothelial cell survival, migration, and proliferation during angiogenesis, is expressed as five splice variants (121, 145, 165, 189, and 206 aminoacids) encoded by a single gene. Although the three shorter isoforms are mainly diffusible, the two longer ones are sequestered in cell membranes after secretion. However, their potential role as true components of the extracellular matrix has not been investigated. We determined that endothelial cells could adhere and spread on VEGF189 and VEGF165, but not on VEGF121. Adhesion was mediated by the alpha3beta1 and alpha(v)beta3 integrins and other alpha(v) integrins but not by the cognate VEGF receptors. Cells migrated on VEGF165 and VEGF189 and displayed a stellate morphology with numerous lamellopodia and FAK staining but no actin stress fibers. Tumstatin, an antiangiogenic peptide that interacts with the alpha(v)beta3 integrin, could inhibit adhesion on VEGF, and this effect was potentiated by anti-alpha(v)beta3 blocking antibody. Immobilized VEGF almost totally abolished endothelial cell apoptosis through interactions with integrins. The inhibition of alpha(v)beta3 engagement with immobilized VEGF by tumstatin inhibited most of its survival activity. We have thus determined a new VEGF receptor-independent role for immobilized VEGF in supporting cell adhesion and survival through interactions with integrins.


Assuntos
Adesão Celular , Movimento Celular , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/fisiologia , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Linfocinas/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fatores de Crescimento Endotelial/metabolismo , Fatores de Crescimento Endotelial/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Integrina alfa3beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Linfocinas/metabolismo , Linfocinas/fisiologia , Modelos Biológicos , Fibras de Estresse/ultraestrutura , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
J Cell Sci ; 115(Pt 22): 4201-14, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12376553

RESUMO

Collagens IV, XV and XVIII are major components of various basement membranes. In addition to the collagen-specific triple helix, these collagens are characterized by the presence of several non-collagenous domains. It is clear now that these ubiquitous collagen molecules are involved in more subtle and sophisticated functions than just the molecular architecture of basement membranes, particularly in the context of extracellular matrix degradation. Degradation of the basement membrane collagens occurs during numerous physiological and pathological processes such as embryonic development or tumorigenesis and generates collagen fragments. These fragments are involved in the regulation of functions differing from those of their original intact molecules. The non-collagenous C-terminal fragment NC1 of collagen IV, XV and XVIII have been recently highlighted in the literature because of their potential in reducing angiogenesis and tumorigenesis, but it is clear that their biological functions are not limited to these processes. Proteolytic release of soluble NC1 fragments stimulates migration, proliferation, apoptosis or survival of different cell types and suppresses various morphogenetic events.


Assuntos
Membrana Basal/metabolismo , Colágeno/metabolismo , Células Eucarióticas/metabolismo , Animais , Membrana Basal/ultraestrutura , Colágeno/genética , Células Eucarióticas/citologia , Humanos , Invasividade Neoplásica/fisiopatologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...