Your browser doesn't support javascript.
loading
LepR+ niche cell-derived AREG compromises hematopoietic stem cell maintenance under conditions of DNA repair deficiency and aging.
Wu, Limei; Lin, Qiqi; Chatla, Srinivas; Amarachintha, Surya; Wilson, Andrew F; Atale, Neha; Gao, Zhenxia J; Joseph, Jonathan; Wolff, Emily V; Du, Wei.
Afiliação
  • Wu L; Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA.
  • Lin Q; Genome Stability Program, UPMC Hillman Cancer Center, Pittsburgh, PA.
  • Chatla S; Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA.
  • Amarachintha S; Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA.
  • Wilson AF; Department of Biology, Georgia Southwestern State University, Americus, GA.
  • Atale N; Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
  • Gao ZJ; Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA.
  • Joseph J; Genome Stability Program, UPMC Hillman Cancer Center, Pittsburgh, PA.
  • Wolff EV; Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA.
  • Du W; Genome Stability Program, UPMC Hillman Cancer Center, Pittsburgh, PA.
Blood ; 142(18): 1529-1542, 2023 11 02.
Article em En | MEDLINE | ID: mdl-37584437
ABSTRACT
The cross talk between extrinsic niche-derived and intrinsic hematopoietic stem cell (HSC) factors controlling HSC maintenance remains elusive. Here, we demonstrated that amphiregulin (AREG) from bone marrow (BM) leptin receptor (LepR+) niche cells is an important factor that mediates the cross talk between the BM niche and HSCs in stem cell maintenance. Mice deficient of the DNA repair gene Brca2, specifically in LepR+ cells (LepR-Cre;Brca2fl/fl), exhibited increased frequencies of total and myeloid-biased HSCs. Furthermore, HSCs from LepR-Cre;Brca2fl/fl mice showed compromised repopulation, increased expansion of donor-derived, myeloid-biased HSCs, and increased myeloid output. Brca2-deficient BM LepR+ cells exhibited persistent DNA damage-inducible overproduction of AREG. Ex vivo treatment of wild-type HSCs or systemic treatment of C57BL/6 mice with recombinant AREG impaired repopulation, leading to HSC exhaustion. Conversely, inhibition of AREG by an anti-AREG-neutralizing antibody or deletion of the Areg gene in LepR-Cre;Brca2fl/fl mice rescued HSC defects caused by AREG. Mechanistically, AREG activated the phosphoinositide 3-kinases (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, promoted HSC cycling, and compromised HSC quiescence. Finally, we demonstrated that BM LepR+ niche cells from other DNA repair-deficient and aged mice also showed persistent DNA damage-associated overexpression of AREG, which exerts similar negative effects on HSC maintenance. Therefore, we identified an important factor that regulates HSCs function under conditions of DNA repair deficiency and aging.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Distúrbios no Reparo do DNA / Receptores para Leptina Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Blood Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Panamá

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Distúrbios no Reparo do DNA / Receptores para Leptina Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Blood Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Panamá