Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Med ; 21(7): 820-6, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26099046

RESUMO

Interleukin-18 (IL18) participates in atherogenesis through several putative mechanisms. Interruption of IL18 action reduces atherosclerosis in mice. Here, we show that absence of the IL18 receptor (IL18r) does not affect atherosclerosis in apolipoprotein E-deficient (Apoe(-/-)) mice, nor does it affect IL18 cell surface binding to or signaling in endothelial cells. As identified initially by co-immunoprecipitation with IL18, we found that IL18 interacts with the Na-Cl co-transporter (NCC; also known as SLC12A3), a 12-transmembrane-domain ion transporter protein preferentially expressed in the kidney. NCC is expressed in atherosclerotic lesions, where it colocalizes with IL18r. In Apoe(-/-) mice, combined deficiency of IL18r and NCC, but not single deficiency of either protein, protects mice from atherosclerosis. Peritoneal macrophages from Apoe(-/-) mice or from Apoe(-/-) mice lacking IL18r or NCC show IL18 binding and induction of cell signaling and cytokine and chemokine expression, but macrophages from Apoe(-/-) mice with combined deficiency of IL18r and NCC have a blunted response. An interaction between NCC and IL18r on macrophages was detected by co-immunoprecipitation. IL18 binds to the cell surface of NCC-transfected COS-7 cells, which do not express IL18r, and induces cell signaling and cytokine expression. This study identifies NCC as an IL18-binding protein that collaborates with IL18r in cell signaling, inflammatory molecule expression, and experimental atherogenesis.


Assuntos
Aterosclerose/metabolismo , Interleucina-18/metabolismo , Receptores de Interleucina-18/metabolismo , Animais , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/patologia , Células COS , Quimiocinas/metabolismo , Chlorocebus aethiops , Macrófagos/metabolismo , Camundongos , Ligação Proteica , Receptores de Interleucina-18/deficiência , Transdução de Sinais , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Túnica Íntima/patologia
2.
EMBO Mol Med ; 6(7): 952-69, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24963147

RESUMO

Immunoglobulin E (IgE) activates mast cells (MCs). It remains unknown whether IgE also activates other inflammatory cells, and contributes to the pathogenesis of abdominal aortic aneurysms (AAAs). This study demonstrates that CD4+ T cells express IgE receptor FcεR1, at much higher levels than do CD8+ T cells. IgE induces CD4+ T-cell production of IL6 and IFN-γ, but reduces their production of IL10. FcεR1 deficiency (Fcer1a-/-) protects apolipoprotein E-deficient (Apoe-/-) mice from angiotensin-II infusion-induced AAAs and reduces plasma IL6 levels. Adoptive transfer of CD4+ T cells (but not CD8+ T cells), MCs, and macrophages from Apoe-/- mice, but not those from Apoe-/- Fcer1a-/- mice, increases AAA size and plasma IL6 in Apoe-/- Fcer1a-/- recipient mice. Biweekly intravenous administration of an anti-IgE monoclonal antibody ablated plasma IgE and reduced AAAs in Apoe-/- mice. Patients with AAAs had significantly higher plasma IgE levels than those without AAAs. This study establishes an important role of IgE in AAA pathogenesis by activating CD4+ T cells, MCs, and macrophages and supports consideration of neutralizing plasma IgE in the therapeutics of human AAAs.


Assuntos
Aneurisma da Aorta Abdominal/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunoglobulina E/imunologia , Macrófagos/imunologia , Mastócitos/imunologia , Transferência Adotiva , Animais , Aneurisma da Aorta Abdominal/sangue , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/patologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Feminino , Deleção de Genes , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/genética , Macrófagos/patologia , Masculino , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de IgE/genética , Receptores de IgE/imunologia , Regulação para Cima
3.
Cardiovasc Res ; 96(3): 543-51, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22871590

RESUMO

AIMS: Mast cells participate importantly in abdominal aortic aneurysms (AAAs) by releasing inflammatory cytokines to promote vascular cell protease expression and arterial wall remodelling. Mast cells accumulate in AAA lesions during disease progression, but the exact chemokines by which mast cells migrate to the site of vascular inflammation remain unknown. This study tested the hypothesis that mast cells use chemokine (C-C motif) receptor 2 (CCR2) for their accumulation in experimental mouse AAA lesions. METHODS AND RESULTS: We generated mast cell and apolipoprotein E double-deficient (Apoe(-/-)Kit(W-sh/W-sh)) mice and found that they were protected from angiotensin II (Ang II) chronic infusion-induced AAAs compared with Apoe(-/-) littermates. Using bone-marrow derived mast cells (BMMC) from Apoe(-/-) mice and CCR2 double-deficient (Apoe(-/-)Ccr2(-/-)) mice, we demonstrated that Apoe(-/-)Kit(W-sh/W-sh) mice receiving BMMC from Apoe(-/-)Ccr2(-/-) mice, but not those from Apoe(-/-) mice, remained protected from AAA formation. Adoptive transfer of BMMC from Apoe(-/-) mice into Apoe(-/-)Kit(W-sh/W-sh) mice also increased lesion content of macrophages, T cells, and MHC class II-positive cells; there was also increased apoptosis, angiogenesis, cell proliferation, elastin fragmentation, and medial smooth muscle cell loss. In contrast, adoptive transfer of BMMC from Apoe(-/-)Ccr2(-/-) mice into Apoe(-/-)Kit(W-sh/W-sh) mice did not affect these variables. CONCLUSIONS: The increased AAA formation and associated lesion characteristics in Apoe(-/-)Kit(W-sh/W-sh) mice after receiving BMMC from Apoe(-/-) mice, but not from Apoe(-/-)Ccr2(-/-) mice, suggests that mast cells use CCR2 as the chemokine receptor for their recruitment in Ang II-induced mouse AAA lesions.


Assuntos
Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/imunologia , Quimiotaxia , Mastócitos/imunologia , Receptores CCR2/metabolismo , Transferência Adotiva , Angiotensina II , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apoptose , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Elastina/metabolismo , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Mastócitos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Neovascularização Patológica , Proteínas Proto-Oncogênicas c-kit/deficiência , Proteínas Proto-Oncogênicas c-kit/genética , Receptores CCR2/deficiência , Receptores CCR2/genética
4.
Front Immunol ; 3: 7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22566893

RESUMO

Mast cells (MCs) play an important role in allergic hyperresponsiveness and in defending microorganism infections. Recent studies of experimental animals and humans have suggested that MCs participate in obesity and diabetes. MC distribution and activities in adipose tissues may vary, depending on the locations of different adipose tissues. In addition to releasing inflammatory mediators to affect adipose tissue extracellular matrix remodeling and to promote inflammatory cell recruitment and proliferation, MCs directly and indirectly interact and activate adipose tissue cells, including adipocytes and recruited inflammatory cells. Plasma MC protease levels are significantly higher in obese patients than in lean subjects. Experimental obese animals lose body weight after MC inactivation. MC functions in diabetes are even more complicated, and depend on the type of diabetes and on different diabetic complications. Both plasma MC proteases and MC activation essential immunoglobulin E levels are significant risk factors for human pre-diabetes and diabetes mellitus. MC stabilization prevents diet-induced diabetes and improves pre-established diabetes in experimental animals. MC depletion or inactivation can improve diet-induced type 2 diabetes and some forms of type 1 diabetes, but also can worsen other forms of type 1 diabetes, at least in experimental animals. Observations from animal and human studies have suggested beneficial effects of treating diabetic patients with MC stabilizers. Some diabetic patients may benefit from enhancing MC survival and proliferation - hypotheses that merit detailed basic researches and clinical studies.

5.
PLoS One ; 5(11): e13973, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21085595

RESUMO

BACKGROUND: Cysteine protease cathepsins are important in extracellular matrix protein degradation, cell apoptosis, and angiogenesis. Mice lacking cathepsins are protected from tumor progression in several animal models, suggesting that the regulation of cathepsin activities controls the growth of various malignant tumors. METHODS AND RESULTS: We tested the role of cathepsins using a mouse model of multistage epithelial carcinogenesis, in which the human keratin-14 promoter/enhancer drove the expression of human papillomavirus type 16 (HPV16) early region E6/E7 transgenes. During the progression of premalignant dysplasia, we observed increased expression of cysteine protease cathepsin S, but concomitantly reduced expression of cathepsin endogenous inhibitor cystatin C in the skin tissue extract. Absence of cystatin C in these transgenic mice resulted in more progression of dysplasia to carcinoma in situ on the face, ear, chest, and tail. Chest and ear skin extract real time PCR and immunoblot analysis, mouse serum sample ELISA, tissue immunohistological analysis, and tissue extract-mediated in vitro elastinolysis and collagenolysis assays demonstrated that cystatin C deficiency significantly increased cathepsin expression and activity. In skin from both the chest and ear, we found that the absence of cystatin C reduced epithelial cell apoptosis but increased proliferation. From the same tissue preparations, we detected significantly higher levels of pro-angiogenic laminin 5-derived γ2 peptides and concurrently increased neovascularization in cystatin C-deficient mice, compared to those from wild-type control mice. CONCLUSION: Enhanced cathepsin expression and activity in cystatin C-deficient mice contributed to the progression of dysplasia by altering premalignant tissue epithelial proliferation, apoptosis, and neovascularization.


Assuntos
Cistatina C/deficiência , Epiderme/metabolismo , Queratina-14/genética , Proteínas Oncogênicas Virais/genética , Animais , Apoptose , Catepsinas/genética , Catepsinas/metabolismo , Proliferação de Células , Cistatina C/genética , Cistatina C/metabolismo , Progressão da Doença , Epiderme/patologia , Feminino , Papillomavirus Humano 16/genética , Humanos , Hiperplasia , Immunoblotting , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas E7 de Papillomavirus/genética , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
6.
Sci Signal ; 3(146): pe40, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-21045204

RESUMO

MicroRNAs (miRNAs) effectively regulate gene expression in cultured cells and human disease models, and such regulation can be blocked with antibodies against miRNAs if miRNA-associated adverse effects occur. Promising findings using miRNAs to prevent disease progression in animal studies give hope to patients with disorders caused by dysregulated gene expression, such as cardiovascular diseases. Inflammatory cell infiltration, endothelial cell dysfunction, and angiogenesis are common pathologies of cardiovascular diseases. Accumulating data suggest that miRNA-mediated inhibition of gene expression can drive these pathologies in cardiac tissue or vasculature. It is often desirable to deliver exogenously prepared miRNAs or antibodies against miRNAs to target genes or miRNAs in specific cell or tissue types. Because naked miRNAs or antibodies against miRNAs are often unstable in the circulation, investigation has focused on their packaging and efficient delivery to diseased organs.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Sistemas de Liberação de Medicamentos , MicroRNAs/administração & dosagem , Animais , Humanos , Camundongos , MicroRNAs/uso terapêutico , Ratos
7.
Arterioscler Thromb Vasc Biol ; 28(12): 2202-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18818416

RESUMO

OBJECTIVE: Previous studies demonstrated increased levels of cysteine proteases cathepsins in serum and adipose tissues from obese patients. We now provide evidence from a mouse model of obesity to suggest a direct participation of cathepsin K (CatK) in mouse body weight gain and glucose metabolism. METHODS AND RESULTS: Using real-time polymerase chain reaction, we detected 12-fold increase in CatK transcripts after adipogenesis of human preadipocytes. Using an immunohistology analysis, we consistently observed high levels of CatK expression in adipose tissues from obese humans and mice. Selective inhibition of CatK activity blocked the lipid accumulation in human and mouse preadipocytes. In mice, CatK deficiency reduced significantly diet-induced body weight gain and serum glucose and insulin levels. Similar results were obtained in diet-induced and genetically created (ob/ob) obese mice after animals were treated with a CatK-selective inhibitor. Mechanistic study demonstrated a role for CatK in degrading fibronectin, a matrix protein that controls adipogenesis. Deficiency or inhibition of CatK leads to fibronectin accumulation in muscle and adipose tissues. CONCLUSIONS: This study demonstrates an essential role of CatK in adipogenesis and mouse body weight gain, possibly via degradation of fibronectin, thus suggesting a novel therapeutic strategy for the control of obesity by regulating CatK activity.


Assuntos
Catepsinas/antagonistas & inibidores , Catepsinas/deficiência , Glucose/metabolismo , Aumento de Peso/fisiologia , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Adipogenia/fisiologia , Animais , Glicemia/metabolismo , Catepsina K , Catepsinas/genética , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Feminino , Fibronectinas/metabolismo , Humanos , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Aumento de Peso/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA