Your browser doesn't support javascript.
loading
Enteroendocrine Progenitor Cell-Enriched miR-7 Regulates Intestinal Epithelial Proliferation in an Xiap-Dependent Manner.
Singh, Ajeet P; Hung, Yu-Han; Shanahan, Michael T; Kanke, Matt; Bonfini, Alessandro; Dame, Michael K; Biraud, Mandy; Peck, Bailey C E; Oyesola, Oyebola O; Freund, John M; Cubitt, Rebecca L; Curry, Ennessa G; Gonzalez, Liara M; Bewick, Gavin A; Tait-Wojno, Elia D; Kurpios, Natasza A; Ding, Shengli; Spence, Jason R; Dekaney, Christopher M; Buchon, Nicolas; Sethupathy, Praveen.
Afiliación
  • Singh AP; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Hung YH; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Shanahan MT; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Kanke M; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Bonfini A; Cornell Institute of Host-Microbe Interactions and Disease. Department of Entomology, Cornell University, New York.
  • Dame MK; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
  • Biraud M; Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina.
  • Peck BCE; Department of Surgery, University of Michigan, Ann Arbor, Michigan.
  • Oyesola OO; Baker Institute of Animal Health and Department of Microbiology and Immunology, Cornell University, Ithaca, New York.
  • Freund JM; Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina.
  • Cubitt RL; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Curry EG; Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina.
  • Gonzalez LM; Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina.
  • Bewick GA; Diabetes Research Group, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.
  • Tait-Wojno ED; Baker Institute of Animal Health and Department of Microbiology and Immunology, Cornell University, Ithaca, New York.
  • Kurpios NA; Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York.
  • Ding S; Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina.
  • Spence JR; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
  • Dekaney CM; Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina.
  • Buchon N; Cornell Institute of Host-Microbe Interactions and Disease. Department of Entomology, Cornell University, New York.
  • Sethupathy P; Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York. Electronic address: pr46@cornell.edu.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Article en En | MEDLINE | ID: mdl-31756561
ABSTRACT
BACKGROUND &

AIMS:

The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis.

METHODS:

We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice.

RESULTS:

First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling.

CONCLUSIONS:

This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.
Asunto(s)
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Células Madre / Células Enteroendocrinas / MicroARNs / Proteínas Inhibidoras de la Apoptosis / Mucosa Intestinal Tipo de estudio: Prognostic_studies Idioma: En Revista: Cell Mol Gastroenterol Hepatol Año: 2020 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Células Madre / Células Enteroendocrinas / MicroARNs / Proteínas Inhibidoras de la Apoptosis / Mucosa Intestinal Tipo de estudio: Prognostic_studies Idioma: En Revista: Cell Mol Gastroenterol Hepatol Año: 2020 Tipo del documento: Article