Your browser doesn't support javascript.
loading
Evidence that the loss of colonic anti-microbial peptides may promote dysbiotic Gram-negative inflammaging-associated bacteria in aging mice.
Forsyth, Christopher B; Shaikh, Maliha; Engen, Phillip A; Preuss, Fabian; Naqib, Ankur; Palmen, Breanna A; Green, Stefan J; Zhang, Lijuan; Bogin, Zlata R; Lawrence, Kristi; Sharma, Deepak; Swanson, Garth R; Bishehsari, Faraz; Voigt, Robin M; Keshavarzian, Ali.
Affiliation
  • Forsyth CB; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.
  • Shaikh M; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Engen PA; Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States.
  • Preuss F; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Naqib A; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Palmen BA; Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States.
  • Green SJ; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Zhang L; Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States.
  • Bogin ZR; Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States.
  • Lawrence K; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Sharma D; Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States.
  • Swanson GR; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Bishehsari F; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Voigt RM; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
  • Keshavarzian A; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
Front Aging ; 5: 1352299, 2024.
Article in En | MEDLINE | ID: mdl-38501032
ABSTRACT

Introduction:

Aging studies in humans and mice have played a key role in understanding the intestinal microbiome and an increased abundance of "inflammaging" Gram-negative (Gn) bacteria. The mechanisms underlying this inflammatory profile in the aging microbiome are unknown. We tested the hypothesis that an aging-related decrease in colonic crypt epithelial cell anti-microbial peptide (AMP) gene expression could promote colonic microbiome inflammatory Gn dysbiosis and inflammaging.

Methods:

As a model of aging, C57BL/6J mice fecal (colonic) microbiota (16S) and isolated colonic crypt epithelial cell gene expression (RNA-seq) were assessed at 2 months (mth) (human 18 years old; yo), 15 mth (human 50 yo), and 25 mth (human 84 yo). Informatics examined aging-related microbial compositions, differential colonic crypt epithelial cell gene expressions, and correlations between colonic bacteria and colonic crypt epithelial cell gene expressions.

Results:

Fecal microbiota exhibited significantly increased relative abundances of pro-inflammatory Gn bacteria with aging. Colonic crypt epithelial cell gene expression analysis showed significant age-related downregulation of key AMP genes that repress the growth of Gn bacteria. The aging-related decrease in AMP gene expressions is significantly correlated with an increased abundance in Gn bacteria (dysbiosis), loss of colonic barrier gene expression, and senescence- and inflammation-related gene expression.

Conclusion:

This study supports the proposed model that aging-related loss of colonic crypt epithelial cell AMP gene expression promotes increased relative abundances of Gn inflammaging-associated bacteria and gene expression markers of colonic inflammaging. These data may support new targets for aging-related therapies based on intestinal genes and microbiomes.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Front Aging Year: 2024 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Front Aging Year: 2024 Document type: Article Affiliation country: United States