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

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Neuroinflammation ; 16(1): 221, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31727092

RESUMO

BACKGROUND: Obese mice on a high-fat diet (HFD) display signs of inflammation in the hypothalamic arcuate nucleus (ARC), a critical area for controlling systemic energy metabolism. This has been suggested as a key mechanism of obesity-associated hypothalamic dysfunction. We reported earlier that bone marrow-derived macrophages accumulate in the ARC to sustain hypothalamic inflammation upon chronic exposure to an HFD. However, the mechanism underlying hypothalamic macrophage accumulation has remained unclear. METHODS: We investigated whether circulating monocytes or myeloid precursors contribute to hypothalamic macrophage expansion during chronic HFD feeding. To trace circulating myeloid cells, we generated mice that express green fluorescent protein (GFP) in their lysozyme M-expressing myeloid cells (LysMGFP mice). We conducted parabiosis and bone marrow transplantation experiments using these animals. Mice received an HFD for 12 or 30 weeks and were then sacrificed to analyze LysMGFP cells in the hypothalamus. Hypothalamic vascular permeability in the HFD-fed obese mice was also tested by examining the extravascular leakage of Evans blue and fluorescence-labeled albumin. The timing of LysMGFP cell entry to the hypothalamus during development was also evaluated. RESULTS: Our parabiosis and bone marrow transplantation experiments revealed a significant infiltration of circulating LysMGFP cells into the liver, skeletal muscle, choroid plexus, and leptomeninges but not in the hypothalamic ARC during chronic HFD feeding, despite increased hypothalamic vascular permeability. These results suggested that the recruitment of circulating monocytes is not a major mechanism for maintaining and expanding the hypothalamic macrophage population in diet-induced obesity. We demonstrated instead that LysMGFP cells infiltrate the hypothalamus during its development. LysMGFP cells appeared in the hypothalamic area from the late embryonic period. This cellular pool suddenly increased immediately after birth, peaked at the postnatal second week, and adopted an adult pattern of distribution after weaning. CONCLUSIONS: Bone marrow-derived macrophages mostly populate the hypothalamus in early postnatal life and may maintain their pool without significant recruitment of circulating monocytes throughout life, even under conditions of chronic HFD feeding.


Assuntos
Hipotálamo/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Animais , Transplante de Medula Óssea , Permeabilidade Capilar , Dieta Hiperlipídica , Metabolismo Energético , Resistência à Insulina/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Parabiose
2.
Arch Craniofac Surg ; 20(4): 246-250, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31462016

RESUMO

Recently, there is a growing interest of hyperbaric oxygen therapy in many fields of medicine. We had a 43-year-old female patient presented with severe necrosis of the nose, philtrum, and upper lip due to retrograde arterial occlusion after nasolabial fold hyaluronic acid filler injection. Our patient went through 43 sessions of systemic hyperbaric oxygen therapy from December 2, 2017 to January 18, 2018. We administered 2.8 atmosphere absolute (ATA) for 135 minutes in the first session and the remaining sessions consisted of 2.0 ATA for 110 minutes. In reporting this case, we wish to provide a warning regarding the latent risk of filler injections and share our experience about minimizing soft tissue damage in the early stages with systemic hyperbaric oxygen therapy.

3.
FEBS Lett ; 591(12): 1742-1751, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28542876

RESUMO

Obesity-induced hypothalamic inflammation is closely associated with various metabolic complications and neurodegenerative disorders. Astrocytes, the most abundant glial cells in the central nervous system, play a crucial role in pathological hypothalamic inflammatory processes. Here, we demonstrate that hypothalamic astrocytes accumulate lipid droplets under saturated fatty acid-rich conditions, such as obese environment, and that the lipid-laden astrocytes increase astrogliosis markers and inflammatory cytokines (TNFα, IL-1ß, IL-6, MCP-1) at the transcript and/or protein level. Medium conditioned by the lipid-laden astrocytes stimulate microglial chemotactic activity and upregulate transcripts of the microglia activation marker Iba-1 and inflammatory cytokines. These findings indicate that the lipid-laden astrocytes formed in free fatty acid-rich obese condition may participate in obesity-induced hypothalamic inflammation through promoting microglia migration and activation.


Assuntos
Astrócitos/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Metabolismo dos Lipídeos , Microglia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/imunologia , Astrócitos/patologia , Biomarcadores/metabolismo , Linhagem Celular , Movimento Celular , Células Cultivadas , Quimiotaxia , Citocinas/genética , Ácidos Graxos não Esterificados/efeitos adversos , Hipotálamo/citologia , Hipotálamo/imunologia , Hipotálamo/patologia , Gotículas Lipídicas/imunologia , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/patologia , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/imunologia , Microglia/patologia , Proteínas do Tecido Nervoso/genética , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/patologia , Ácido Palmítico/efeitos adversos , RNA Mensageiro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA