Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 32(12): 3012-23, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23087364

RESUMO

OBJECTIVE: Activation of inflammatory pathways plays a critical role in the development of abdominal aortic aneurysms (AAA). Notch1 signaling is a significant regulator of the inflammatory response; however, its role in AAA is unknown. METHODS AND RESULTS: In an angiotensin II-induced mouse model of AAA, activation of Notch1 signaling was observed in the aortic aneurysmal tissue of Apoe(-/-) mice, and a similar activation of Notch1 was observed in aneurysms of humans undergoing AAA repair. Notch1 haploinsufficiency significantly reduced the incidence of AAA in Apoe(-/-) mice in response to angiotensin II. Reconstitution of bone marrow-derived cells from Notch1(+/-);Apoe(-/-) mice (donor) in lethally irradiated Apoe(-/-) mice (recipient) decreased the occurrence of aneurysm. Flow cytometry and immunohistochemistry demonstrated that Notch1 haploinsufficiency prevented the influx of inflammatory macrophages at the aneurysmal site by causing defects in macrophage migration and proliferation. In addition, there was an overall reduction in the inflammatory burden in the aorta of the Notch1(+/-);Apoe(-/-) mice compared with the Apoe(-/-) mice. Last, pharmacological inhibition of Notch1 signaling also prevented AAA formation and progression in Apoe(-/-) mice. CONCLUSIONS: Our data suggest that decreased levels of Notch1 protect against the formation of AAA by preventing macrophage recruitment and attenuating the inflammatory response in the aorta.


Assuntos
Aneurisma da Aorta Abdominal/prevenção & controle , Arterite/prevenção & controle , Macrófagos/fisiologia , Receptor Notch1/deficiência , Receptor Notch1/genética , Transdução de Sinais/fisiologia , Angiotensina II/efeitos adversos , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/fisiopatologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Arterite/fisiopatologia , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Haploinsuficiência/genética , Humanos , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Receptor Notch1/fisiologia , Transdução de Sinais/efeitos dos fármacos
2.
PLoS One ; 12(5): e0178538, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28562688

RESUMO

AIMS: Infiltration of macrophages and apoptosis of vascular smooth muscle cells (VSMCs) promote the development of abdominal aortic aneurysm (AAA). Previously, we demonstrated that global Notch1 deficiency prevents the formation of AAA in a mouse model. Herein, we sought to explore the cell-specific roles of Notch1 in AAA development. METHODS AND RESULTS: Cell-specific Notch1 haploinsufficient mice, generated on Apoe-/- background using Cre-lox technology, were infused with angiotensin II (1000 ng/min/kg) for 28 days. Notch1 haploinsufficiency in myeloid cells (n = 9) prevented the formation of AAA attributed to decreased inflammation. Haploinsufficiency of Notch1 in SMCs (n = 14) per se did not prevent AAA formation, but histoarchitectural traits of AAA including elastin degradation and aortic remodeling, were minimal in SMC-Notch1+/-;Apoe-/- mice compared to Apoe-/- mice (n = 33). Increased immunostaining of the contractile SMC-phenotype markers and concomitant decreased expression of synthetic SMC-phenotype markers were observed in the aortae of SMC-Notch1+/-;Apoe-/- mice. Expression of connective tissue growth factor (CTGF), a matrix-associated protein that modulates the synthetic VSMC phenotype, increased in the abdominal aorta of Apoe-/- mice and in the adventitial region of the abdominal aorta in human AAA. Notch1 haploinsufficiency decreased the expression of Ctgf in the aorta and in vitro cell culture system. In vitro studies on SMCs using the Notch1 intracellular domain (NICD) plasmid, dominant negative mastermind-like (dnMAML), or specific siRNA suggest that Notch1, not Notch3, directly modulates the expression of CTGF. CONCLUSIONS: Our data suggest that lack of Notch1 in SMCs limits dilation of the abdominal aorta by maintaining contractile SMC-phenotype and preventing matrix-remodeling.


Assuntos
Aneurisma da Aorta Abdominal/patologia , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Haploinsuficiência , Músculo Liso Vascular/metabolismo , Receptor Notch1/metabolismo , Animais , Aneurisma da Aorta Abdominal/metabolismo , Células Cultivadas , Técnicas de Cocultura , Metaloproteinases da Matriz/biossíntese , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia
3.
J Am Heart Assoc ; 3(6): e001064, 2014 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-25349182

RESUMO

BACKGROUND: The progression of abdominal aortic aneurysm (AAA) involves a sustained influx of proinflammatory macrophages, which exacerbate tissue injury by releasing cytokines, chemokines, and matrix metalloproteinases. Previously, we showed that Notch deficiency reduces the development of AAA in the angiotensin II-induced mouse model by preventing infiltration of macrophages. Here, we examined whether Notch inhibition in this mouse model prevents progression of small AAA and whether these effects are associated with altered macrophage differentiation. METHODS AND RESULTS: Treatment with pharmacological Notch inhibitor (DAPT [N-(N-[3,5-difluorophenacetyl]-L-alanyl)-S-phenylglycine t-butyl ester]) at day 3 or 8 of angiotensin II infusion arrested the progression of AAA in Apoe(-/-) mice, as demonstrated by a decreased luminal diameter and aortic width. The abdominal aortas of Apoe(-/-) mice treated with DAPT showed decreased expression of matrix metalloproteinases and presence of elastin precursors including tropoelastin and hyaluronic acid. Marginal adventitial thickening observed in the aorta of DAPT-treated Apoe(-/-) mice was not associated with increased macrophage content, as observed in the mice treated with angiotensin II alone. Instead, DAPT-treated abdominal aortas showed increased expression of Cd206-positive M2 macrophages and decreased expression of Il12-positive M1 macrophages. Notch1 deficiency promoted M2 differentiation of macrophages by upregulating transforming growth factor ß2 in bone marrow-derived macrophages at basal levels and in response to IL4. Protein expression of transforming growth factor ß2 and its downstream effector pSmad2 also increased in DAPT-treated Apoe(-/-) mice, indicating a potential link between Notch and transforming growth factor ß2 signaling in the M2 differentiation of macrophages. CONCLUSIONS: Pharmacological inhibitor of Notch signaling prevents the progression of AAA by macrophage differentiation-dependent mechanisms. The study also provides insights for novel therapeutic strategies to prevent the progression of small AAA.


Assuntos
Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/tratamento farmacológico , Dipeptídeos/farmacologia , Receptor Notch1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Idoso , 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 , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Elastina/metabolismo , Regulação da Expressão Gênica , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos Knockout , Pessoa de Meia-Idade , Receptor Notch1/deficiência , Receptor Notch1/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA