1
|
Wang W, Hariharan M, Ding W, Bartlett A, Barragan C, Castanon R, Rothenberg V, Song H, Nery J, Aldridge A, Altshul J, Kenworthy M, Liu H, Tian W, Zhou J, Zeng Q, Chen H, Wei B, Gündüz IB, Norell T, Broderick TJ, McClain MT, Satterwhite LL, Burke TW, Petzold EA, Shen X, Woods CW, Fowler VG, Ruffin F, Panuwet P, Barr DB, Beare JL, Smith AK, Spurbeck RR, Vangeti S, Ramos I, Nudelman G, Sealfon SC, Castellino F, Walley AM, Evans T, Müller F, Greenleaf WJ, Ecker JR. Genetics and Environment Distinctively Shape the Human Immune Cell Epigenome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.29.546792. [PMID: 37425926 PMCID: PMC10327221 DOI: 10.1101/2023.06.29.546792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The epigenomic landscape of human immune cells is dynamically shaped by both genetic factors and environmental exposures. However, the relative contributions of these elements are still not fully understood. In this study, we employed single-nucleus methylation sequencing and ATAC-seq to systematically explore how pathogen and chemical exposures, along with genetic variation, influence the immune cell epigenome. We identified distinct exposure-associated differentially methylated regions (eDMRs) corresponding to each exposure, revealing how environmental factors remodel the methylome, alter immune cell states, and affect transcription factor binding. Furthermore, we observed a significant correlation between changes in DNA methylation and chromatin accessibility, underscoring the coordinated response of the epigenome. We also uncovered genotype-associated DMRs (gDMRs), demonstrating that while eDMRs are enriched in regulatory regions, gDMRs are preferentially located in gene body marks, suggesting that exposures and genetic factors exert differential regulatory control. Notably, disease-associated SNPs were frequently colocalized with meQTLs, providing new cell-type-specific insights into the genetic basis of disease. Our findings underscore the intricate interplay between genetic and environmental factors in sculpting the immune cell epigenome, offering a deeper understanding of how immune cell function is regulated in health and disease.
Collapse
Affiliation(s)
- Wenliang Wang
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Manoj Hariharan
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Wubin Ding
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Cesar Barragan
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Rosa Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Vince Rothenberg
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Haili Song
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Joseph Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Andrew Aldridge
- Duke University School of Medicine, Bryan Research Building, 311 Research Drive, Durham, NC 27710, USA
| | - Jordan Altshul
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Mia Kenworthy
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Wei Tian
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jingtian Zhou
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Qiurui Zeng
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Huaming Chen
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Bei Wei
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Irem B. Gündüz
- Integrative Cellular Biology & Bioinformatics Lab, Saarland University, 66123 Saarbrücken, Germany
| | - Todd Norell
- Healthspan, Resilience, and Performance, Florida Institute for Human and Machine Cognition, 40 S Alcaniz St, Pensacola, FL 32502, USA
| | - Timothy J Broderick
- Healthspan, Resilience, and Performance, Florida Institute for Human and Machine Cognition, 40 S Alcaniz St, Pensacola, FL 32502, USA
| | - Micah T. McClain
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
- Durham Veterans Affairs Medical Center, Durham, NC 27705 USA
| | - Lisa L. Satterwhite
- Department of Civil and Environmental Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Thomas W. Burke
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
| | - Elizabeth A. Petzold
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Christopher W. Woods
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
- Durham Veterans Affairs Medical Center, Durham, NC 27705 USA
| | - Vance G. Fowler
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
- Duke Clinical Research Institute, Durham NC 27701 USA
| | - Felicia Ruffin
- Center for Infectious Disease Diagnostics and Innovation, Division of Infectious Diseases, Duke University Medical Center, Durham, NC 27710 USA
| | - Parinya Panuwet
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322 USA
| | - Dana B. Barr
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322 USA
| | | | - Anthony K. Smith
- Battelle Memorial Institute, 505 King Ave Columbus OH 43201, USA
| | | | - Sindhu Vangeti
- Department of Neurology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Irene Ramos
- Department of Neurology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - German Nudelman
- Department of Neurology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Stuart C. Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Flora Castellino
- U.S. Department of Health and Human Services, Administration for Strategic Preparedness and Response, Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Anna Maria Walley
- Vaccitech plc, Unit 6-10, Zeus Building, Rutherford Avenue, Harwell OX11 0DF, United Kingdom
| | - Thomas Evans
- Vaccitech plc, Unit 6-10, Zeus Building, Rutherford Avenue, Harwell OX11 0DF, United Kingdom
| | - Fabian Müller
- Integrative Cellular Biology & Bioinformatics Lab, Saarland University, 66123 Saarbrücken, Germany
| | | | - Joseph R. Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
2
|
Khan NM, Wilderman A, Kaiser JM, Kamalakar A, Goudy SL, Cotney J, Drissi H. Enhanced osteogenic potential of iPSC-derived mesenchymal progenitor cells following genome editing of GWAS variants in the RUNX1 gene. Bone Res 2024; 12:70. [PMID: 39643619 PMCID: PMC11624199 DOI: 10.1038/s41413-024-00369-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 12/09/2024] Open
Abstract
Recent genome-wide association studies (GWAS) identified 518 significant loci associated with bone mineral density (BMD), including variants at the RUNX1 locus (rs13046645, rs2834676, and rs2834694). However, their regulatory impact on RUNX1 expression and bone formation remained unclear. This study utilized human induced pluripotent stem cells (iPSCs) differentiated into osteoblasts to investigate these variants' regulatory roles. CRISPR/Cas9 was employed to generate mutant (Δ) iPSC lines lacking these loci at the RUNX1 locus. Deletion lines (Δ1 and Δ2) were created in iPSCs to assess the effects of removing regions containing these loci. Deletion lines exhibited enhanced osteogenic potential, with increased expression of osteogenic marker genes and Alizarin Red staining. Circularized chromosome conformation capture (4C-Seq) was utilized to analyze interactions between BMD-associated loci and the RUNX1 promoter during osteogenesis. Analysis revealed altered chromatin interactions with multiple gene promoters including RUNX1 isoform, as well as SETD4, a histone methyltransferase, indicating their regulatory influence. Interestingly, both deletion lines notably stimulated the expression of the long isoform of RUNX1, with more modest effects on the shorter isoform. Consistent upregulation of SETD4 and other predicted targets within the Δ2 deletion suggested its removal removed a regulatory hub constraining expression of multiple genes at this locus. In vivo experiments using a bone defect model in mice demonstrated increased bone regeneration with homozygous deletion of the Δ2 region. These findings indicate that BMD-associated variants within the RUNX1 locus regulate multiple effector genes involved in osteoblast commitment, providing valuable insights into genetic regulation of bone density and potential therapeutic targets.
Collapse
Affiliation(s)
- Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Andrea Wilderman
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Jarred M Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Archana Kamalakar
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven L Goudy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA.
- Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
3
|
Plotkin LI, Bruzzaniti A, Pianeta R. Sexual Dimorphism in the Musculoskeletal System: Sex Hormones and Beyond. J Endocr Soc 2024; 8:bvae153. [PMID: 39309123 PMCID: PMC11413583 DOI: 10.1210/jendso/bvae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Indexed: 09/25/2024] Open
Abstract
Mounting evidence indicates that whereas some fundamental aspects of bone cell differentiation and function are similar in females and males, there is a clear contribution of sex/gender on the effects of signaling molecules on bone mass and strength and, consequently, on the effects of pharmacologic approaches to treat skeletal disorders. However, until recently, most studies were designed and performed using only 1 sex, resulting in a scarcity of published information on sexual dimorphism of the musculoskeletal system, including the mandible/masticatory muscles and the axial and appendicular bones and skeletal muscles. Further, it is now recognized that scientific rigor requires the study of both males and females. Therefore, there is an increasing need to understand the molecular and cellular basis for the differential outcomes of genetic manipulations and therapeutic agent administration depending on the sex of the experimental animals. Studies have shown higher muscle mass, cancellous bone mass, and long bone width in males compared with females as well as different traits in the pelvis and the skull, which are usually used for gender identification in forensic anthropology. Yet, most reports focus on the role of sex hormones, in particular, the consequences of estrogen deficiency with menopause in humans and in ovariectomized animal models. In addition, emerging data is starting to unveil the effects of gender-affirming hormonal therapy on the musculoskeletal system. We summarize here the current knowledge on the sex/gender-dependent phenotypic characteristics of the bone and skeletal muscles in humans and rodents, highlighting studies in which side by side comparisons were made.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Angela Bruzzaniti
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN 46202-5120, USA
| | - Roquelina Pianeta
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202-5120, USA
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| |
Collapse
|
4
|
Siewe N, Friedman A. Osteoporosis induced by cellular senescence: A mathematical model. PLoS One 2024; 19:e0303978. [PMID: 38805428 PMCID: PMC11132490 DOI: 10.1371/journal.pone.0303978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Abstract
Osteoporosis is a disease characterized by loss of bone mass, where bones become fragile and more likely to fracture. Bone density begins to decrease at age 50, and a state of osteoporosis is defined by loss of more than 25%. Cellular senescence is a permanent arrest of normal cell cycle, while maintaining cell viability. The number of senescent cells increase with age. Since osteoporosis is an aging disease, it is natural to consider the question to what extend senescent cells induce bone density loss and osteoporosis. In this paper we use a mathematical model to address this question. We determine the percent of bone loss for men and women during age 50 to 100 years, and the results depend on the rate η of net formation of senescent cell, with η = 1 being the average rate. In the case η = 1, the model simulations are in agreement with empirical data. We also consider senolytic drugs, like fisetin and quercetin, that selectively eliminate senescent cells, and assess their efficacy in terms of reducing bone loss. For example, at η = 1, with estrogen hormonal therapy and early treatment with fisetin, bone density loss for women by age 75 is 23.4% (below osteoporosis), while with no treatment with fisetin it is 25.8% (osteoporosis); without even a treatment with estrogen hormonal therapy, bone loss of 25.3% occurs already at age 65.
Collapse
Affiliation(s)
- Nourridine Siewe
- School of Mathematics and Statistics, College of Science, Rochester Institute of Technology, Rochester, New York, United States of America
| | - Avner Friedman
- Department of Mathematics, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
5
|
潘 子, 周 雪, 曹 志, 潘 剑. [Latest Findings on the Role of RUNX1 in Bone Development and Disorders]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:256-262. [PMID: 38645858 PMCID: PMC11026898 DOI: 10.12182/20240360103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Indexed: 04/23/2024]
Abstract
Runt-related transcription factor (RUNX1) is a transcription factor closely involved in hematopoiesis. RUNX1 gene mutation plays an essential pathogenic role in the initiation and development of hematological tumors, especially in acute myeloid leukemia. Recent studies have shown that RUNX1 is also involved in the regulation of bone development and the pathological progression of bone-related diseases. RUNX1 promotes the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts and modulates the maturation and extracellular matrix formation of chondrocytes. The expression of RUNX1 in mesenchymal stem cells, chondrocytes, and osteoblasts is of great significance for maintaining normal bone development and the mass and quality of bones. RUNX1 also inhibits the differentiation and bone resorptive activities of osteoclasts, which may be influenced by sexual dimorphism. In addition, RUNX1 deficiency contributes to the pathogenesis of osteoarthritis, delayed fracture healing, and osteoporosis, which was revealed by the RUNX1 conditional knockout modeling in mice. However, the roles of RUNX1 in regulating the hypertrophic differentiation of chondrocytes, the sexual dimorphism of activities of osteoclasts, as well as bone loss in diabetes mellitus, senescence, infection, chronic inflammation, etc, are still not fully understood. This review provides a systematic summary of the research progress concerning RUNX1 in the field of bone biology, offering new ideas for using RUNX1 as a potential target for bone related diseases, especially osteoarthritis, delayed fracture healing, and osteoporosis.
Collapse
Affiliation(s)
- 子建 潘
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 口腔颌面外科 (成都 610041)State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - 雪儿 周
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 口腔颌面外科 (成都 610041)State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - 志炜 曹
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 口腔颌面外科 (成都 610041)State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - 剑 潘
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 口腔颌面外科 (成都 610041)State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
6
|
Lv Y, Yao X, Ling Q, Suo S, Wang J, Zhao S, Gao X, Tong H, Jin J, Zhang X, Yu W. Osteolytic lesion as initial presentation in FIP1L1-PDGFRA-rearranged myeloid/lymphoid neoplasm with eosinophilia: a case report. Ann Hematol 2024; 103:357-360. [PMID: 37777636 DOI: 10.1007/s00277-023-05485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023]
Affiliation(s)
- Yunfei Lv
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - XingYun Yao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, 310030, Hangzhou, China
| | - Qing Ling
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Shanshan Suo
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Jinghan Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Shuqi Zhao
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, 310030, Hangzhou, China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China.
| | - Xiang Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China.
| | - Wenjuan Yu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Rd, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Key Laboratory of Hematologic Malignancy, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Hematological disorders, Hangzhou, 310003, Zhejiang, People's Republic of China.
- Zhejiang University Cancer Center, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
7
|
Desai S, Lång P, Näreoja T, Windahl SH, Andersson G. RANKL-dependent osteoclast differentiation and gene expression in bone marrow-derived cells from adult mice is sexually dimorphic. Bone Rep 2023; 19:101697. [PMID: 37485233 PMCID: PMC10359713 DOI: 10.1016/j.bonr.2023.101697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023] Open
Abstract
Sex-specific differences in bone integrity and properties are associated with age as well as the number and activity of cells involved in bone remodeling. The aim of this study was to investigate sex-specific differences in adhesion, proliferation, and differentiation of mouse bone marrow derived cells into osteoclasts. The adherent fraction of bone marrow- derived cells from 12-week-old male and female C57BL/6J mice were assessed for their adhesion, proliferation, and receptor activator of nuclear factor κB (RANKL)-induced differentiation into osteoclasts. Female bone marrow derived macrophages (BMDMs) displayed higher adhesion and proliferation ratio upon macrophage colony stimulating factor (M-CSF) (day 0) and M-CSF + RANKL (day 4) treatment, respectively. On the contrary, male BMDMs differentiated more efficiently into osteoclasts upon RANKL-treatment compared to females (day 5). To further understand these sex-specific differences at the gene expression level, BMDMs treated with M-CSF (day 0) and M-CSF + RANKL (day 4), were assessed for their differential expression of genes through RNA sequencing. M-CSF treatment resulted in 1106 differentially expressed genes, while RANKL-treatment gave 473 differentially expressed genes. Integrin, adhesion, and proliferation-associated genes were elevated in the M-CSF-treated female BMDMs. RANKL-treatment further enhanced the expression of the proliferation- associated genes, and of genes associated with inhibition of osteoclast differentiation in the females, while RANK-signaling-associated genes were upregulated in males. In conclusion, BMDM adhesion, proliferation and differentiation into osteoclasts are sex-specific and may be directed by the PI3K-Akt signaling pathway for proliferation, and the colony stimulating factor 1-receptor and the RANKLsignaling pathway for the differentiation.
Collapse
Affiliation(s)
- Suchita Desai
- Karolinska Institutet, Department of Laboratory Medicine - Division of Pathology, Huddinge, Sweden
| | - Pernilla Lång
- Karolinska Institutet, Department of Laboratory Medicine - Division of Pathology, Huddinge, Sweden
| | - Tuomas Näreoja
- Karolinska Institutet, Department of Laboratory Medicine - Division of Pathology, Huddinge, Sweden
- Department of Life Technologies, University of Turku, Finland
| | - Sara H. Windahl
- Karolinska Institutet, Department of Laboratory Medicine - Division of Pathology, Huddinge, Sweden
| | - Göran Andersson
- Karolinska Institutet, Department of Laboratory Medicine - Division of Pathology, Huddinge, Sweden
| |
Collapse
|
8
|
Xie L, Feng E, Li S, Chai H, Chen J, Li L, Ge J. Comparisons of gene expression between peripheral blood mononuclear cells and bone tissue in osteoporosis. Medicine (Baltimore) 2023; 102:e33829. [PMID: 37335694 PMCID: PMC10194530 DOI: 10.1097/md.0000000000033829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/02/2023] [Indexed: 06/21/2023] Open
Abstract
Osteoporosis (OP) is one of the major public health problems in the world. However, the biomarkers between the peripheral blood mononuclear cells (PBMs) and bone tissue for prognosis of OP have not been well characterized. This study aimed to explore the similarities and differences of the gene expression profiles between the PBMs and bone tissue and identify potential genes, transcription factors (TFs) and hub proteins involved in OP. The patients were enrolled as an experimental group, and healthy subjects served as normal controls. Human whole-genome expression chips were used to analyze gene expression profiles from PBMs and bone tissue. And the differentially expressed genes (DEGs) were subsequently studied using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. The above DEGs were constructed into protein-protein interaction network. Finally, TF-DEGs regulation networks were constructed. Microarray analysis revealed that 226 DEGs were identified between OP and normal controls in the PBMs, while 2295 DEGs were identified in the bone tissue. And 13 common DEGs were obtained by comparing the 2 tissues. The Gene Ontology analysis indicated that DEGs in the PBMs were more involved in immune response, while DEGs in bone were more involved in renal response and urea transmembrane transport. And the Kyoto Encyclopedia of Genes and Genomes analysis indicated almost all of the pathways in the PBMs were overlapped with those in the bone tissue. Furthermore, protein-protein interaction network presented 6 hub proteins: PI3K1, APP, GNB5, FPR2, GNG13, and PLCG1. APP has been found to be associated with OP. Finally, 5 key TFs were identified by TF-DEGs regulation networks analysis (CREB1, RUNX1, STAT3, CREBBP, and GLI1) and were supposed to be associated with OP. This study enhanced our understanding of the pathogenesis of OP. PI3K1, GNB5, FPR2, GNG13, and PLCG1 might be the potential targets of OP.
Collapse
Affiliation(s)
- Lihua Xie
- Key Research Laboratory of Osteoporosis Syndrome Genomics, Fujian Academy of Chinese Medical Sciences, Fuzhou, China
| | - Eryou Feng
- Department of Arthrosis Surgery, Fuzhou Second Hospital, Fuzhou, China
| | - Shengqiang Li
- Key Research Laboratory of Osteoporosis Syndrome Genomics, Fujian Academy of Chinese Medical Sciences, Fuzhou, China
| | - Hao Chai
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Juan Chen
- Key Research Laboratory of Osteoporosis Syndrome Genomics, Fujian Academy of Chinese Medical Sciences, Fuzhou, China
| | - Li Li
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jirong Ge
- Key Research Laboratory of Osteoporosis Syndrome Genomics, Fujian Academy of Chinese Medical Sciences, Fuzhou, China
| |
Collapse
|
9
|
Liu H, Zhai L, Liu Y, Lu D, Vander Ark A, Yang T, Krawczyk CM. The histone demethylase KDM5C controls female bone mass by promoting energy metabolism in osteoclasts. SCIENCE ADVANCES 2023; 9:eadg0731. [PMID: 37018401 PMCID: PMC10075994 DOI: 10.1126/sciadv.adg0731] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 05/28/2023]
Abstract
Women experience osteoporosis at higher rates than men. Aside from hormones, the mechanisms driving sex-dependent bone mass regulation are not well understood. Here, we demonstrate that the X-linked H3K4me2/3 demethylase KDM5C regulates sex-specific bone mass. Loss of KDM5C in hematopoietic stem cells or bone marrow monocytes increases bone mass in female but not male mice. Mechanistically, loss of KDM5C impairs the bioenergetic metabolism, resulting in impaired osteoclastogenesis. Treatment with the KDM5 inhibitor reduces osteoclastogenesis and energy metabolism of both female mice and human monocytes. Our report details a sex-dependent mechanism for bone homeostasis, connecting epigenetic regulation to osteoclast metabolism and positions KDM5C as a potential target for future treatment of osteoporosis in women.
Collapse
Affiliation(s)
- Huadie Liu
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Lukai Zhai
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ye Liu
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Di Lu
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Alexandra Vander Ark
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Tao Yang
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Connie M. Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
10
|
The histone demethylase KDM5C controls female bone mass by promoting energy metabolism in osteoclasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529728. [PMID: 36865269 PMCID: PMC9980061 DOI: 10.1101/2023.02.23.529728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Women experience osteoporosis at higher rates than men. Aside from hormones, the mechanisms driving sex-dependent bone mass regulation are not well-understood. Here, we demonstrate that the X-linked H3K4me2/3 demethylase KDM5C regulates sex-specific bone mass. Loss of KDM5C in hematopoietic stem cells or bone marrow monocytes (BMM) increases bone mass in female but not male mice. Mechanistically, loss of KDM5C impairs the bioenergetic metabolism resulting in impaired osteoclastogenesis. Treatment with the KDM5 inhibitor reduces osteoclastogenesis and energy metabolism of both female mice and human monocytes. Our report details a novel sex-dependent mechanism for bone homeostasis, connecting epigenetic regulation to osteoclast metabolism, and positions KDM5C as a target for future treatment of osteoporosis in women. One-Sentence Summary KDM5C, an X-linked epigenetic regulator, controls female bone homeostasis by promoting energy metabolism in osteoclasts.
Collapse
|
11
|
Breast Cancer Exosomal microRNAs Facilitate Pre-Metastatic Niche Formation in the Bone: A Mathematical Model. Bull Math Biol 2023; 85:12. [PMID: 36607440 DOI: 10.1007/s11538-022-01117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/26/2022] [Indexed: 01/07/2023]
Abstract
Pre-metastatic niche is a location where cancer cells, separating from a primary tumor, find "fertile soil" for growth and proliferation, ensuring successful metastasis. Exosomal miRNAs of breast cancer are known to enter the bone and degrade it, which facilitates cancer cells invasion into the bone interior and ensures its successful colonization. In this paper, we use a mathematical model to first describe, in health, the continuous remodeling of the bone by bone-forming osteoblasts, bone-resorbing osteoclasts and the RANKL-OPG-RANK signaling system, which keeps the balance between bone formation and bone resorption. We next demonstrate how breast cancer exosomal miRNAs disrupt this balance, either by increasing or by decreasing the ratio of osteoclasts/osteoblasts, which results in abnormal high bone resorption or abnormal high bone forming, respectively, and in bone weakening in both cases. Finally we consider the case of abnormally high resorption and evaluate the effect of drugs, which may increase bone density to normal level, thus protecting the bone from invasion by cancer cells.
Collapse
|
12
|
Yang B, Qin Y, Zhang A, Wang P, Jiang H, Shi Y, You G, Shen D, Ni S, Guo L, Liu Y. Circular RNA CircSLC8A1 contributes to osteogenic differentiation in hBMSCs via CircSLC8A1/miR-144-3p/RUNX1 in periprosthetic osteolysis. J Cell Mol Med 2022; 27:189-203. [PMID: 36541023 PMCID: PMC9843530 DOI: 10.1111/jcmm.17633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Circular RNAs (circRNAs) are often found in eukaryocyte and have a role in the pathogenesis of a variety of human disorders. Our related research has shown the differential expression of circRNAs in periprosthetic osteolysis (PPOL). However, the involvement of circRNAs in the exact process is yet unknown. CircSLC8A1 expression was evaluated in clinical samples and human bone marrow mesenchymal stem cells (hBMSCs) in this investigation using quantitative real-time PCR. In vitro and in vivo studies were conducted to explicate its functional role and pathway. We demonstrated CircSLC8A1 is involved in PPOL using gain- and loss-of-function methods. The association of CircSLC8A1 and miR-144-3p, along with miR-144-3p and RUNX1, was predicted using bioinformatics. RNA pull-down and luciferase assays confirmed it. The impact of CircSLC8A1 in the PPOL-mouse model was also investigated using adeno-associated virus. CircSLC8A1 was found to be downregulated in PPOL patients' periprosthetic tissues. Overexpression of CircSLC8A1 promoted osteogenic differentiation (OD) and inhibited apoptosis of hBMSCs in vitro. The osteogenic markers of RUNX1, osteopontin (OPN) and osteocalcin (OCN) were significantly upregulated in hBMSCs after miR-144-3p inhibitor was transferred. Mechanistic analysis demonstrated that CircSLC8A1 directly bound to miR-144-3p and participated in PPOL through the miR-144-3p/RUNX1 pathway in hBMSCs. Micro-CT and quantitative analysis showed that CircSLC8A1 markedly inhibited PPOL, and osteogenic markers (RUNX1, OPN and OCN) were significantly increased (P<0.05) in the mice model. Our findings prove that CircSLC8A1 exerted a regulatory role in promoting osteogenic differentiation in hBMSCs, and CircSLC8A1/miR-144-3p/RUNX1 pathway may provide a potential target for prevention of PPOL.
Collapse
Affiliation(s)
- Boning Yang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Yu Qin
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Ao Zhang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Penghao Wang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Hua Jiang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Yunyi Shi
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Guanchao You
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Dianlin Shen
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Shenghui Ni
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Lei Guo
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Ying Liu
- Department of Nursing, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| |
Collapse
|
13
|
Díaz-Hernández ME, Kinter CW, Watson SR, Mella-Velazquez G, Kaiser J, Liu G, Khan NM, Roberts JL, Lorenzo J, Drissi H. Sexually Dimorphic Increases in Bone Mass Following Tissue-specific Overexpression of Runx1 in Osteoclast Precursors. Endocrinology 2022; 163:6650061. [PMID: 35880727 PMCID: PMC9337273 DOI: 10.1210/endocr/bqac113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Indexed: 11/19/2022]
Abstract
Many metabolic bone diseases arise as a result excessive osteoclastic bone resorption, which has motivated efforts to identify new molecular targets that can inhibit the formation or activity of these bone-resorbing cells. Mounting evidence indicates that the transcription factor Runx1 acts as a transcriptional repressor of osteoclast formation. Prior studies using a conditional knockout approach suggested that Runx1 in osteoclast precursors acts as an inhibitor of osteoclastogenesis; however, the effects of upregulation of Runx1 on osteoclast formation remain unknown. In this study, we investigated the skeletal effects of conditional overexpression of Runx1 in preosteoclasts by crossing novel Runx1 gain-of-function mice (Rosa26-LSL-Runx1) with LysM-Cre transgenic mice. We observed a sex-dependent effect whereby overexpression of Runx1 in female mice increased trabecular bone microarchitectural indices and improved torsion biomechanical properties. These effects were likely mediated by delayed osteoclastogenesis and decreased bone resorption. Transcriptomics analyses during osteoclastogenesis revealed a distinct transcriptomic profile in the Runx1-overexpressing cells, with enrichment of genes related to redox signaling, apoptosis, osteoclast differentiation, and bone remodeling. These data further confirm the antiosteoclastogenic activities of Runx1 and provide new insight into the molecular targets that may mediate these effects.
Collapse
Affiliation(s)
| | | | - Shana R Watson
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Giovanni Mella-Velazquez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Jarred Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Guanglu Liu
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Joseph L Roberts
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia 30329, USA
- Atlanta VA Health Care System, Decatur, Georgia, 30033, USA
| | - Joseph Lorenzo
- Department of Medicine, UConn Health, Farmington, 06032, Connecticut, USA
| | - Hicham Drissi
- Correspondence: Hicham Drissi, PhD, Department of Orthopaedics, Emory University School of Medicine, 21 Ortho Ln, 6th Fl, Office 12, Atlanta, GA 30329, USA.
| |
Collapse
|
14
|
Mun SH, Jastrzebski S, Kalinowski J, Zeng S, Oh B, Bae S, Eugenia G, Khan NM, Drissi H, Zhou P, Shin B, Lee S, Lorenzo J, Park‐Min K. Sexual Dimorphism in Differentiating Osteoclast Precursors Demonstrates Enhanced Inflammatory Pathway Activation in Female Cells. J Bone Miner Res 2021; 36:1104-1116. [PMID: 33567098 PMCID: PMC11140852 DOI: 10.1002/jbmr.4270] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Sexual dimorphism of the skeleton is well documented. At maturity, the male skeleton is typically larger and has a higher bone density than the female skeleton. However, the underlying mechanisms for these differences are not completely understood. In this study, we examined sexual dimorphism in the formation of osteoclasts between cells from female and male mice. We found that the number of osteoclasts in bones was greater in females. Similarly, in vitro osteoclast differentiation was accelerated in female osteoclast precursor (OCP) cells. To further characterize sex differences between female and male osteoclasts, we performed gene expression profiling of cultured, highly purified, murine bone marrow OCPs that had been treated for 3 days with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). We found that 125 genes were differentially regulated in a sex-dependent manner. In addition to genes that are contained on sex chromosomes, transcriptional sexual dimorphism was found to be mediated by genes involved in innate immune and inflammatory response pathways. Furthermore, the NF-κB-NFATc1 axis was activated earlier in female differentiating OCPs, which partially explains the differences in transcriptomic sexual dimorphism in these cells. Collectively, these findings identify multigenic sex-dependent intrinsic difference in differentiating OCPs, which results from an altered response to osteoclastogenic stimulation. In humans, these differences could contribute to the lower peak bone mass and increased risk of osteoporosis that females demonstrate relative to males. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Se Hwan Mun
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Sandra Jastrzebski
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Judy Kalinowski
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Steven Zeng
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Seyeon Bae
- Department of Medicine Weill Cornell Medical College New York NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Giannopoulou Eugenia
- Biological Sciences Department New York City College of Technology, City University of New York Brooklyn NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| | - Nazir M Khan
- Department of Orthopaedics School of Medicine, Emory University Atlanta GA USA
| | - Hicham Drissi
- Department of Orthopaedics School of Medicine, Emory University Atlanta GA USA
| | - Ping Zhou
- Feil Family Brain & Mind Research Institute (BMRI), Weill Cornell Medical College New York NY USA
| | - Bongjin Shin
- Center on Aging University of Connecticut Health Farmington CT USA
| | - Sun‐Kyeong Lee
- Center on Aging University of Connecticut Health Farmington CT USA
| | - Joseph Lorenzo
- Department of Orthopaedic Surgery University of Connecticut Health Farmington CT USA
- Department of Medicine University of Connecticut Health Farmington CT USA
| | - Kyung‐Hyun Park‐Min
- BCMB Allied Program Weill Cornell Graduate School of Medical Sciences New York NY USA
- Department of Medicine Weill Cornell Medical College New York NY USA
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center Hospital for Special Surgery New York NY USA
| |
Collapse
|
15
|
Ling W, Krager K, Richardson KK, Warren AD, Ponte F, Aykin-Burns N, Manolagas SC, Almeida M, Kim HN. Mitochondrial Sirt3 contributes to the bone loss caused by aging or estrogen deficiency. JCI Insight 2021; 6:146728. [PMID: 33878033 PMCID: PMC8262324 DOI: 10.1172/jci.insight.146728] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/14/2021] [Indexed: 12/20/2022] Open
Abstract
Altered mitochondria activity in osteoblasts and osteoclasts has been implicated in the loss of bone mass associated with aging and estrogen deficiency — the 2 most common causes of osteoporosis. However, the mechanisms that control mitochondrial metabolism in bone cells during health or disease remain unknown. The mitochondrial deacetylase sirtuin-3 (Sirt3) has been earlier implicated in age-related diseases. Here, we show that deletion of Sirt3 had no effect on the skeleton of young mice but attenuated the age-related loss of bone mass in both sexes. This effect was associated with impaired bone resorption. Osteoclast progenitors from aged Sirt3-null mice were able to differentiate into osteoclasts, though the differentiated cells exhibited impaired polykaryon formation and resorptive activity, as well as decreased oxidative phosphorylation and mitophagy. The Sirt3 inhibitor LC-0296 recapitulated the effects of Sirt3 deletion in osteoclast formation and mitochondrial function, and its administration to aging mice increased bone mass. Deletion of Sirt3 also attenuated the increase in bone resorption and loss of bone mass caused by estrogen deficiency. These findings suggest that Sirt3 inhibition and the resulting impairment of osteoclast mitochondrial function could be a novel therapeutic intervention for the 2 most important causes of osteoporosis.
Collapse
Affiliation(s)
- Wen Ling
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kimberly K Richardson
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine
| | - Aaron D Warren
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Filipa Ponte
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stavros C Manolagas
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Maria Almeida
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology, Department of Internal Medicine.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
16
|
Abstract
Osteoclasts are the principal mediators of bone resorption. They form through the fusion of mononuclear precursor cells under the principal influence of the cytokines macrophage colony stimulating factor (M-CSF, aka CSF-1) and receptor activator of NF-κB ligand (RANKL, aka TNFSF11). Sexual dimorphism in the development of the skeleton and in the incidence of skeletal diseases is well described. In general, females, at any given age, have a lower bone mass than males. The reasons for the differences in the bone mass of the skeleton between women and men at various ages, and the incidence of certain metabolic bone diseases, are multitude, and include the actions of sex steroids, genetics, age, environment and behavior. All of these influence the rate that osteoclasts form, resorb and die, and frequently produce different effects in females and males. Hence, a variety of factors are responsible for the sexual dimorphism of the skeleton and the activity of osteoclasts in bone. This review will provide an overview of what is currently known about these factors and their effects on osteoclasts.
Collapse
|
17
|
Network Analysis Identifies Gene Regulatory Network Indicating the Role of RUNX1 in Human Intervertebral Disc Degeneration. Genes (Basel) 2020; 11:genes11070771. [PMID: 32659941 PMCID: PMC7397129 DOI: 10.3390/genes11070771] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/31/2022] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD) is a multifactorial physiological process which is often associated with lower back pain. Previous studies have identified some molecular markers associated with disc degeneration, which despite their significant contributions, have provided limited insight into the etiology of IDD. In this study, we utilized a network medicine approach to uncover potential molecular mediators of IDD. Our systematic analyses of IDD associated with 284 genes included functional annotation clustering, interaction networks, network cluster analysis and Transcription factors (TFs)-target gene network analysis. The functional enrichment and protein–protein interaction network analysis highlighted the role of inflammatory genes and cytokine/chemokine signaling in IDD. Moreover, sub-network analysis identified significant clusters possessing organized networks of 24 cytokine and chemokine genes, which may be considered as key modulators for IDD. The expression of these genes was validated in independent microarray datasets. In addition, the regulatory network analysis identified the role of multiple transcription factors, with RUNX1 being a master regulator in the pathogenesis of IDD. Our analyses highlighted the role of cytokine genes and interacting pathways in IDD and further improved our understanding of the genetic mechanisms underlying IDD.
Collapse
|
18
|
Paglia DN, Diaz-Hernandez ME, Roberts JL, Kalinowski J, Lorenzo J, Drissi H. Deletion of Runx1 in osteoclasts impairs murine fracture healing through progressive woven bone loss and delayed cartilage remodeling. J Orthop Res 2020; 38:1007-1015. [PMID: 31769548 DOI: 10.1002/jor.24537] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/24/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023]
Abstract
Conditional deletion of the transcription factor Runt-related transcription factor 1 (Runx1) in myeloid osteoclast precursors promotes osteoclastogenesis and subsequent bone loss. This study posits whether Runx1 regulates clastic cell-mediated bone and cartilage resorption in the fracture callus. We first generated mice, in which Runx1 was conditionally abrogated in osteoclast precursors (LysM-Cre;Runx1F/F ; Runx1 cKO). Runx1 cKO and control mice were then subjected to experimental mid-diaphyseal femoral fractures. Our study found differential resorption of bony and calcified cartilage callus matrix by osteoclasts and chondroclasts within Runx1 cKO calluses, with increased early bony callus resorption and delayed calcified cartilage resorption. There was an increased number of osteoclasts and chondroclasts in the chondro-osseous junction of Runx1 cKO calluses starting at day 11 post-fracture, with minimal woven bone occupying the callus at day 18 post-fracture. LysM-Cre;Runx1F/F mutant mice had increased bone compliance at day 28, but their strength and work to failure were comparable with controls. Taken together, these results indicate that Runx1 is a critical transcription factor in controlling osteoclastogenesis that negatively regulates bone and cartilage resorption in the fracture callus. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:1007-1015, 2020.
Collapse
Affiliation(s)
- David N Paglia
- Department of Orthopaedics, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | | | - Joseph L Roberts
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, Georgia
| | - Judy Kalinowski
- Department of Medicine and Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Joseph Lorenzo
- Department of Medicine and Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Hicham Drissi
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
Tian X, Ma J, Wang T, Tian J, Zheng Y, Peng R, Wang Y, Zhang Y, Mao L, Xu H, Wang S. Long non-coding RNA RUNXOR accelerates MDSC-mediated immunosuppression in lung cancer. BMC Cancer 2018; 18:660. [PMID: 29914443 PMCID: PMC6006856 DOI: 10.1186/s12885-018-4564-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
Background RUNX1 overlapping RNA (RUNXOR) is a long non-coding RNA that has been indicated as a key regulator in the development of myeloid cells by targeting runt-related transcription factor 1 (RUNX1). Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells consisting of immature granulocytes and monocytes with immunosuppression. However, the impact of lncRNA RUNXOR on the development of MDSCs remains unknown. Methods Both the expressions of RUNXOR and RUNX1 in the peripheral blood were measured by qRT-PCR. Human MDSCs used in this study were isolated from tumor tissue of patients with lung cancer by FCM or induced from PBMCs of healthy donors with IL-1β + GM-CSF. Specific siRNA was used to knockdown the expression of RUNXOR in MDSCs. Results In this study, we found that the lncRNA RUNXOR was upregulated in the peripheral blood of lung cancer patients. In addition, as a target gene of RUNXOR, the expression of RUNX1 was downregulated in lung cancer patients. Finally, the expression of RUNXOR was higher in MDSCs isolated from the tumor tissues of lung cancer patients compared with cells from adjacent tissue. In addition, RUNXOR knockdown decreased Arg1 expression in MDSCs. Conclusions Based on our findings, it is illustrated that RUNXOR is significantly associated with the immunosuppression induced by MDSCs in lung cancer patients and may be a target of anti-tumor therapy.
Collapse
Affiliation(s)
- Xinyu Tian
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212012, China.,Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Ma
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ting Wang
- Department of Laboratory Medicine, Jiangsu Cancer Hospital, Nanjing, China
| | - Jie Tian
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yu Zheng
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Rongrong Peng
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yungang Wang
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Zhang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212012, China
| | - Lingxiang Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212012, China.
| | - Huaxi Xu
- Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212012, China. .,Institute of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Growing evidence supports the critical role of transcriptional mechanisms in promoting the spatial and temporal progression of bone healing. In this review, we evaluate and discuss new transcriptional and post-transcriptional regulatory mechanisms of secondary bone repair, along with emerging evidence for epigenetic regulation of fracture healing. RECENT FINDINGS Using the candidate gene approach has identified new roles for several transcription factors in mediating the reactive, reparative, and remodeling phases of fracture repair. Further characterization of the different epigenetic controls of fracture healing and fracture-driven transcriptome changes between young and aged fracture has identified key biological pathways that may yield therapeutic targets. Furthermore, exogenously delivered microRNA to post-transcriptionally control gene expression is quickly becoming an area with great therapeutic potential. Activation of specific transcriptional networks can promote the proper progression of secondary bone healing. Targeting these key factors using small molecules or through microRNA may yield effective therapies to enhance and possibly accelerate fracture healing.
Collapse
Affiliation(s)
- Joseph L Roberts
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, GA, USA
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - David N Paglia
- Department of Orthopaedics, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Hicham Drissi
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, GA, USA.
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA.
- Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA, 30033, USA.
| |
Collapse
|
21
|
Liu Y, Zuo G, Meng X, Gao X, Zhang L, Tang P. Adrenomedullin inhibits osteoclast differentiation through the suppression of receptor activator of nuclear factor-κB ligand-induced nuclear factor-κB activation in glucocorticoid-induced osteoporosis. Exp Ther Med 2017; 14:4009-4016. [PMID: 29067096 PMCID: PMC5647721 DOI: 10.3892/etm.2017.5025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 04/21/2017] [Indexed: 12/22/2022] Open
Abstract
The current study aimed to improve the understanding on the association between adrenomedullin and osteoporosis in mice with glucocorticoid-induced osteoporosis. Bone resorption and osteoporosis-associated indexes, including maximum load, stiffness, energy to failure, ultimate strength, elastic modulus, post-yield displacement and post-yield displacement, in mice with osteoporosis were analyzed in order to evaluate the effect of adrenomedullin. The receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation was investigated subsequent to treatment with adrenomedullin in vitro. The results demonstrated that adrenomedullin significantly improved bone mass loss, density, bone strength and osteoporosis disease in the mice with glucocorticoid-induced osteoporosis. In addition, adrenomedullin markedly improved the osteoporosis-associated NFATc1, TRAP, OSCAR and c-Fos expression levels. Furthermore, the current findings indicated that RANKL-mediated osteoclast differentiation was suppressed in vitro and in vivo. Notably, the data revealed that adrenomedullin significantly improved the osteoporotic symptoms through inhibition of RANKL-induced NF-κB activation in glucocorticoid-induced osteoporosis. In conclusion, adrenomedullin serves an essential role in the progression of glucocorticoid-induced osteoporosis, regulating the bone mass loss, density and strength through the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuanxin Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Guilai Zuo
- Department of Orthopaedics, Pingyin People Hospital, Jinan, Shandong 250400, P.R. China
| | - Xin Meng
- Department of Orthopaedics, Pingyin People Hospital, Jinan, Shandong 250400, P.R. China
| | - Xingxiao Gao
- Department of Orthopaedics, Pingyin People Hospital, Jinan, Shandong 250400, P.R. China
| | - Lihai Zhang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Peifu Tang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
22
|
Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol Rev 2017; 97:1295-1349. [DOI: 10.1152/physrev.00036.2016] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Tomoki Nakashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masahiro Shinohara
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Noriko Komatsu
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinichiro Sawa
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takeshi Nitta
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
23
|
Yang ZQ, Zhang HL, Duan CC, Geng S, Wang K, Yu HF, Yue ZP, Guo B. IGF1 regulates RUNX1 expression via IRS1/2: Implications for antler chondrocyte differentiation. Cell Cycle 2017; 16:522-532. [PMID: 28055425 DOI: 10.1080/15384101.2016.1274471] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although IGF1 is important for the proliferation and differentiation of chondrocytes, its underlying molecular mechanism is still unknown. Here we addressed the physiologic function of IGF1 in antler cartilage and explored the interplay of IGF1, IRS1/2 and RUNX1 in chondrocyte differentiation. The results showed that IGF1 was highly expressed in antler chondrocytes. Exogenous rIGF1 could increase the proliferation of chondrocytes and cell proportion in the S phase, whereas IGF1R inhibitor PQ401 abrogated the induction by rIGF1. Simultaneously, IGF1 could stimulate the expression of IHH which was a well-known marker for prehypertrophic chondrocytes. Further analysis evidenced that IGF1 regulated the expression of IRS1/2 whose silencing resulted in a rise of IHH mRNA levels, but the regulation was impeded by PQ401. Knockdown of IRS1 or IRS2 with specific siRNA could greatly enhance rIGF1-induced chondrocyte differentiation and reduce the expression of RUNX1. Extraneous rRUNX1 might rescue the effects of IRS1 or IRS2 siRNA on the differentiation. In antler chondrocytes, IGF1 played a role in modulating the expression of RUNX1 through IGF1R. Moreover, attenuation of RUNX1 expression advanced the differentiation elicited by rIGF1, while administration of rRUNX1 to chondrocytes treated with IGF1 siRNA or PQ401 reduced their differentiation. Additionally, siRNA-mediated downregulation of IRS1 or IRS2 in the chondrocytes impaired the interaction between IGF1 and RUNX1. Collectively, IGF1 could promote the proliferation and differentiation of antler chondrocytes. Furthermore, IRS1/2 might act downstream of IGF1 to regulate chondrocyte differentiation through targeting RUNX1.
Collapse
Affiliation(s)
- Zhan-Qing Yang
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Hong-Liang Zhang
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Cui-Cui Duan
- b Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences , Changchun , P. R. China
| | - Shuang Geng
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Kai Wang
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Hai-Fan Yu
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Zhan-Peng Yue
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| | - Bin Guo
- a College of Veterinary Medicine, Jilin University , Changchun , P. R. China
| |
Collapse
|