Your browser doesn't support javascript.
loading
Diet-microbiome interactions promote enteric nervous system resilience following spinal cord injury.
Hamilton, Adam M; Blackmer-Raynolds, Lisa; Li, Yaqing; Kelly, Sean; Kebede, Nardos; Williams, Anna; Chang, Jianjun; Garraway, Sandra M; Srinivasan, Shanthi; Sampson, Timothy R.
Afiliación
  • Hamilton AM; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Blackmer-Raynolds L; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Li Y; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Kelly S; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Kebede N; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Williams A; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Chang J; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Garraway SM; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
  • Srinivasan S; Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta GA 30329.
  • Sampson TR; Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30329.
bioRxiv ; 2024 Jun 08.
Article en En | MEDLINE | ID: mdl-38895207
ABSTRACT
Spinal cord injury (SCI) results in a plethora of physiological dysfunctions across all body systems, including intestinal dysmotility and atrophy of the enteric nervous system (ENS). Typically, the ENS has capacity to recover from perturbation, so it is unclear why intestinal pathophysiologies persist after traumatic spinal injury. With emerging evidence demonstrating SCI-induced alterations to the gut microbiome composition, we hypothesized that modulation of the gut microbiome could contribute to enteric nervous system recovery after injury. Here, we show that intervention with the dietary fiber, inulin prevents ENS atrophy and limits SCI-induced intestinal dysmotility in mice. However, SCI-associated microbiomes and exposure to specific SCI-sensitive gut microbes are not sufficient to modulate injury-induced intestinal dysmotility. Intervention with microbially-derived short-chain fatty acid (SCFA) metabolites prevents ENS dysfunctions and phenocopies inulin treatment in injured mice, implicating these microbiome metabolites in protection of the ENS. Notably, inulin-mediated resilience is dependent on signaling by the cytokine IL-10, highlighting a critical diet-microbiome-immune axis that promotes ENS resilience following SCI. Overall, we demonstrate that diet and microbially-derived signals distinctly impact recovery of the ENS after traumatic spinal injury. This protective diet-microbiome-immune axis may represent a foundation to uncover etiological mechanisms and future therapeutics for SCI-induced neurogenic bowel.

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article
...