Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nature ; 552(7684): 248-252, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29211719

RESUMO

Diabetic retinopathy is an important cause of blindness in adults, and is characterized by progressive loss of vascular cells and slow dissolution of inter-vascular junctions, which result in vascular leakage and retinal oedema. Later stages of the disease are characterized by inflammatory cell infiltration, tissue destruction and neovascularization. Here we identify soluble epoxide hydrolase (sEH) as a key enzyme that initiates pericyte loss and breakdown of endothelial barrier function by generating the diol 19,20-dihydroxydocosapentaenoic acid, derived from docosahexaenoic acid. The expression of sEH and the accumulation of 19,20-dihydroxydocosapentaenoic acid were increased in diabetic mouse retinas and in the retinas and vitreous humour of patients with diabetes. Mechanistically, the diol targeted the cell membrane to alter the localization of cholesterol-binding proteins, and prevented the association of presenilin 1 with N-cadherin and VE-cadherin, thereby compromising pericyte-endothelial cell interactions and inter-endothelial cell junctions. Treating diabetic mice with a specific sEH inhibitor prevented the pericyte loss and vascular permeability that are characteristic of non-proliferative diabetic retinopathy. Conversely, overexpression of sEH in the retinal Müller glial cells of non-diabetic mice resulted in similar vessel abnormalities to those seen in diabetic mice with retinopathy. Thus, increased expression of sEH is a key determinant in the pathogenesis of diabetic retinopathy, and inhibition of sEH can prevent progression of the disease.


Assuntos
Retinopatia Diabética/enzimologia , Retinopatia Diabética/prevenção & controle , Epóxido Hidrolases/antagonistas & inibidores , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Modelos Animais de Doenças , Progressão da Doença , Ácidos Docosa-Hexaenoicos/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Ependimogliais , Ácidos Graxos Insaturados/metabolismo , Feminino , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/patologia , Presenilina-1/metabolismo , Retina/efeitos dos fármacos , Retina/enzimologia , Retina/metabolismo , Retina/patologia , Solubilidade , Corpo Vítreo/metabolismo
2.
Prostaglandins Other Lipid Mediat ; 133: 93-102, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28442442

RESUMO

Polyunsaturated fatty acids (PUFA) and their cytochrome P450 (CYP450) metabolites have been linked to angiogenesis and vessel homeostasis. However, the role of individual CYP isoforms and their endogenous metabolites in those processes are not clear. Here, we focused on the role of Cyp2c44 in postnatal retinal angiogenesis and report that Cyp2c44 is highly expressed in Müller glial cells in the retina. The constitutive as well as inducible postnatal genetic deletion of Cyp2c44 resulted in an increased vessel network density without affecting vessel radial expansion during the first postnatal week. This phenotype was associated with an increased endothelial cell proliferation and attenuated Notch signaling. LC-MS/MS analyses revealed that levels of hydroxydocosahexaenoic acids (HDHA), i.e., 10-, 17- and 20-HDHA were significantly elevated in retinas from 5day old Cyp2c44-/- mice compared to their wild-type littermates. Enzymatic activity assays revealed that HDHAs were potential substrates for Cyp2c44 which could account for the increased levels of HDHAs in retinas from Cyp2c44-/- mice. These data indicate that Cyp2c44 is expressed in the murine retina and, like the soluble epoxide hydrolase, is expressed in Müller glia cells. The enhanced endothelial cell proliferation and Notch inhibition seen in retinas from Cyp2c44-deficient mice indicate a role for Cyp2c44-derived lipid mediators in physiological angiogenesis.


Assuntos
Família 2 do Citocromo P450/metabolismo , Células Ependimogliais/enzimologia , Neovascularização Fisiológica , Retina/fisiologia , Animais , Proliferação de Células , Família 2 do Citocromo P450/deficiência , Família 2 do Citocromo P450/genética , Ácidos Docosa-Hexaenoicos/metabolismo , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Retina/citologia
3.
J Exp Med ; 214(9): 2695-2713, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28739604

RESUMO

Metastasis is the primary cause of cancer death. The inflammatory tumor microenvironment contributes to metastasis, for instance, by recruiting blood and lymph vessels. Among tumor-infiltrating immune cells, tumor-associated macrophages (TAMs) take a center stage in promoting both tumor angiogenesis and metastatic spread. We found that genetic deletion of the S1P receptor 1 (S1pr1) alone in CD11bhi CD206+ TAMs infiltrating mouse breast tumors prevents pulmonary metastasis and tumor lymphangiogenesis. Reduced lymphangiogenesis was also observed in the nonrelated methylcholanthrene-induced fibrosarcoma model. Transcriptome analysis of isolated TAMs from both entities revealed reduced expression of the inflammasome component Nlrp3 in S1PR1-deficient TAMs. Macrophage-dependent lymphangiogenesis in vitro was triggered upon inflammasome activation and required both S1PR1 signaling and IL-1ß production. Finally, NLRP3 expression in tumor-infiltrating macrophages correlated with survival, lymph node invasion, and metastasis of mammary carcinoma patients. Conceptually, our study indicates an unappreciated role of the NLRP3 inflammasome in promoting metastasis via the lymphatics downstream of S1PR1 signaling in macrophages.


Assuntos
Interleucina-1beta/fisiologia , Linfangiogênese/fisiologia , Macrófagos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Metástase Neoplásica/fisiopatologia , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Feminino , Fibrossarcoma/fisiopatologia , Humanos , Metástase Linfática , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Esfingosina-1-Fosfato
4.
Sci Signal ; 9(434): ra64, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27353364

RESUMO

Tumor cell-derived factors skew macrophages toward a tumor-supporting phenotype associated with the secretion of protumorigenic mediators. Apoptosing tumor cells release sphingosine 1-phosphate (S1P), which stimulates the production of lipocalin 2 (LCN2) in tumor-associated macrophages and is associated with tumor metastasis. We explored the mechanism by which S1P induces LCN2 in macrophages and investigated how this contributed to tumor growth and metastasis. Knockdown of S1P receptor 1 (S1PR1) in primary human macrophages and experiments with bone marrow-derived macrophages from S1PR1-deficient mice showed that S1P signaled through S1PR1 to induce LCN2 expression. The LCN2 promoter contains a consensus sequence for signal transducer and activator of transcription 3 (STAT3), and deletion of the STAT3 recognition sequence reduced expression of an LCN2-controlled reporter gene. Conditioned medium from coculture experiments indicated that the release of LCN2 from macrophages induced tube formation and proliferation in cultures of primary human lymphatic endothelial cells in a manner dependent on the kinase PI3K and subsequent induction of the growth factor VEGFC, which functioned as an autocrine signal stimulating the receptor VEGFR3. Knockout of Lcn2 attenuated tumor-associated lymphangiogenesis and breast tumor metastasis both in the breast cancer model MMTV-PyMT mice and in mice bearing orthotopic wild-type tumors. Our findings indicate that macrophages respond to dying tumor cells by producing signals that promote lymphangiogenesis, which enables metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Lipocalina-2/metabolismo , Linfangiogênese , Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Lipocalina-2/genética , Lisofosfolipídeos , Células MCF-7 , Macrófagos/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Esfingosina/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA