Your browser doesn't support javascript.
loading
Diet-induced metabolic and immune impairments are sex-specifically modulated by soluble TNF signaling in the 5xFAD mouse model of Alzheimer's disease.
Rodrigues, Maria Elizabeth De Sousa; Bolen, MacKenzie L; Blackmer-Raynolds, Lisa; Schwartz, Noah; Chang, Jianjun; Tansey, Malú Gámez; Sampson, Timothy Robert.
Afiliación
  • Rodrigues MES; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
  • Bolen ML; Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, FL, USA.
  • Blackmer-Raynolds L; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
  • Schwartz N; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
  • Chang J; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
  • Tansey MG; Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, FL, USA; Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA. Electronic address: mgtansey@ufl.e
  • Sampson TR; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA. Electronic address: trsamps@emory.edu.
Neurobiol Dis ; 196: 106511, 2024 Jun 15.
Article en En | MEDLINE | ID: mdl-38670277
ABSTRACT
Emerging evidence indicates that high-fat, high carbohydrate diet (HFHC) impacts central pathological features of Alzheimer's disease (AD) across both human incidences and animal models. However, the mechanisms underlying this association are poorly understood. Here, we identify compartment-specific metabolic and inflammatory dysregulations that are induced by HFHC diet in the 5xFAD mouse model of AD pathology. We observe that both male and female 5xFAD mice display exacerbated adiposity, cholesterolemia, and dysregulated insulin signaling. Independent of biological sex, HFHC diet also resulted in altered inflammatory cytokine profiles across the gastrointestinal, circulating, and central nervous systems (CNS) compartments demonstrating region-specific impacts of metabolic inflammation. Interestingly, inhibiting the inflammatory cytokine, soluble tumor necrosis factor (TNF) with the brain-permeant soluble TNF inhibitor XPro1595 was able to restore aspects of HFHC-induced metabolic inflammation, but only in male mice. Targeted transcriptomics of CNS regions revealed that inhibition of soluble TNF was sufficient to alter expression of hippocampal and cortical genes associated with beneficial immune and metabolic responses. Collectively, these results suggest that HFHC diet impairs metabolic and inflammatory pathways in an AD-relevant genotype and that soluble TNF has sex-dependent roles in modulating these pathways across anatomical compartments. Modulation of energy homeostasis and inflammation may provide new therapeutic avenues for AD.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Ratones Transgénicos / Factor de Necrosis Tumoral alfa / Modelos Animales de Enfermedad / Enfermedad de Alzheimer / Dieta Alta en Grasa Límite: Animals Idioma: En Revista: Neurobiol Dis Asunto de la revista: NEUROLOGIA Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Ratones Transgénicos / Factor de Necrosis Tumoral alfa / Modelos Animales de Enfermedad / Enfermedad de Alzheimer / Dieta Alta en Grasa Límite: Animals Idioma: En Revista: Neurobiol Dis Asunto de la revista: NEUROLOGIA Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos
...