Your browser doesn't support javascript.
loading
Melatonin enhances osteoblastogenesis of senescent bone marrow stromal cells through NSD2-mediated chromatin remodelling.
Xie, Ying; Han, Na; Li, Feng; Wang, Lijuan; Liu, Gerui; Hu, Meilin; Wang, Sheng; Wei, Xuelei; Guo, Jing; Jiang, Hongmei; Wang, Jingjing; Li, Xin; Wang, Yixuan; Wang, Jingya; Bian, Xiyun; Zhu, Zhongjiao; Zhang, Hui; Liu, Chunhua; Liu, Xiaozhi; Liu, Zhiqiang.
Afiliación
  • Xie Y; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Han N; Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital; National Center for Trauma Medicine, Beijing, China.
  • Li F; Department of Orthopaedics, Weifang People's Hospital, Weifang, China.
  • Wang L; Central Laboratory; Linyi Key Laboratory of Tumor Biology, Linyi People's Hospital, Linyi, China.
  • Liu G; Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Hu M; Tianjin Medical University School of Stomatology, Heping, China.
  • Wang S; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Wei X; Department of Emergency, Tianjin Hospital, Tianjin, China.
  • Guo J; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Jiang H; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Wang J; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Li X; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Wang Y; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Wang J; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
  • Bian X; Central Laboratory; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Binhai, Tianjin, China.
  • Zhu Z; Department of Orthopaedics, Tengzhou Central People's Hospital, Tenghzou, China.
  • Zhang H; Department of Cardiology, Heart Centre; Ministry of Education Key Laboratory of Child Development and Disorders National Clinical Research Center for Child Health and Disorders; Chongqing Key Laboratory of Pediatrics; China International Science and Technology Cooperation Base of Child Development a
  • Liu C; Department of Physiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong, China.
  • Liu X; Central Laboratory; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Binhai, Tianjin, China.
  • Liu Z; The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases; Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, China.
Clin Transl Med ; 12(2): e746, 2022 02.
Article en En | MEDLINE | ID: mdl-35220680
ABSTRACT

BACKGROUND:

Aging-associated osteoporosis is frequently seen in the elderly in clinic, but efficient managements are limited because of unclear nosogenesis. The current study aims to investigate the role of melatonin on senescent bone marrow stromal cells (BMSCs) and the underlying regulating mechanism.

METHODS:

Melatonin levels were tested by ELISA. Gene expression profiles were performed by RNA-sequencing, enrichment of H3K36me2 on gene promoters was analyzed by Chromatin Immunoprecipitation Sequencing (ChIP-seq), and chromatin accessibility was determined by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Osteogenesis of BMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining, and in vivo effects of melatonin was assessed by histological staining and micro computed tomography (micro-CT) scan. Correlation of NSD2 expression and severity of senile osteoporosis patients were analyzed by Pearson correlation.

RESULTS:

Melatonin levels were decreased during aging in human bone marrow, accompanied by downregulation of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2) expression in the senescent BMSCs. Melatonin stimulated the expression of NSD2 through MT1/2-mediated signaling pathways, resulting in the rebalancing of H3K36me2 and H3K27me3 modifications to increase chromatin accessibility of the osteogenic genes, runt-related transcription factor 2 (RUNX2) and bone gamma-carboxyglutamate protein (BGLAP). Melatonin promoted osteogenesis of BMSCs in vitro, and alleviates osteoporosis progression in the aging mice. In clinic, severity of senile osteoporosis (SOP) was negatively correlated with melatonin level in bone marrow, as well as NSD2 expression in BMSCs. Similarly, melatonin remarkably enhanced osteogenic differentiation of BMSCs derived from SOP patients in vitro.

CONCLUSIONS:

Collectively, our study dissects previously unreported mechanistic insights into the epigenetic regulating machinery of melatonin in meliorating osteogenic differentiation of senescent BMSC, and provides evidence for application of melatonin in preventing aging-associated bone loss.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Osteoblastos / Proteínas Represoras / N-Metiltransferasa de Histona-Lisina / Ensamble y Desensamble de Cromatina / Células Madre Mesenquimatosas / Melatonina Tipo de estudio: Prognostic_studies Límite: Adolescent / Adult / Aged / Aged80 / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Clin Transl Med Año: 2022 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Osteoblastos / Proteínas Represoras / N-Metiltransferasa de Histona-Lisina / Ensamble y Desensamble de Cromatina / Células Madre Mesenquimatosas / Melatonina Tipo de estudio: Prognostic_studies Límite: Adolescent / Adult / Aged / Aged80 / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Clin Transl Med Año: 2022 Tipo del documento: Article País de afiliación: China