Your browser doesn't support javascript.
loading
Microglial cell dysregulation in brain aging and neurodegeneration.
von Bernhardi, Rommy; Eugenín-von Bernhardi, Laura; Eugenín, Jaime.
Afiliação
  • von Bernhardi R; Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile.
  • Eugenín-von Bernhardi L; Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile.
  • Eugenín J; Laboratory of Neural Systems, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile (USACH) Santiago, Chile.
Front Aging Neurosci ; 7: 124, 2015.
Article em En | MEDLINE | ID: mdl-26257642
Aging is the main risk factor for neurodegenerative diseases. In aging, microglia undergoes phenotypic changes compatible with their activation. Glial activation can lead to neuroinflammation, which is increasingly accepted as part of the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). We hypothesize that in aging, aberrant microglia activation leads to a deleterious environment and neurodegeneration. In aged mice, microglia exhibit an increased expression of cytokines and an exacerbated inflammatory response to pathological changes. Whereas LPS increases nitric oxide (NO) secretion in microglia from young mice, induction of reactive oxygen species (ROS) predominates in older mice. Furthermore, there is accumulation of DNA oxidative damage in mitochondria of microglia during aging, and also an increased intracellular ROS production. Increased ROS activates the redox-sensitive nuclear factor kappa B, which promotes more neuroinflammation, and can be translated in functional deficits, such as cognitive impairment. Mitochondria-derived ROS and cathepsin B, are also necessary for the microglial cell production of interleukin-1ß, a key inflammatory cytokine. Interestingly, whereas the regulatory cytokine TGFß1 is also increased in the aged brain, neuroinflammation persists. Assessing this apparent contradiction, we have reported that TGFß1 induction and activation of Smad3 signaling after inflammatory stimulation are reduced in adult mice. Other protective functions, such as phagocytosis, although observed in aged animals, become not inducible by inflammatory stimuli and TGFß1. Here, we discuss data suggesting that mitochondrial and endolysosomal dysfunction could at least partially mediate age-associated microglial cell changes, and, together with the impairment of the TGFß1-Smad3 pathway, could result in the reduction of protective activation and the facilitation of cytotoxic activation of microglia, resulting in the promotion of neurodegenerative diseases.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Risk_factors_studies Idioma: En Revista: Front Aging Neurosci Ano de publicação: 2015 Tipo de documento: Article País de afiliação: Chile País de publicação: Suíça

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Risk_factors_studies Idioma: En Revista: Front Aging Neurosci Ano de publicação: 2015 Tipo de documento: Article País de afiliação: Chile País de publicação: Suíça