1
|
Jung S, Ha J, Park JH, Yoo KH. Decoding SPP1 regulation: Genetic and nongenetic insights into its role in disease progression. Mol Cells 2025; 48:100215. [PMID: 40210132 PMCID: PMC12049823 DOI: 10.1016/j.mocell.2025.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/18/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Secreted phosphoprotein 1 (SPP1), also known as osteopontin, is a multifunctional glycoprotein that plays a critical role in various physiological processes, including cell adhesion, chemotaxis, immune regulation, and tissue remodeling. Originally identified as a key component of the bone matrix, SPP1 is now recognized for its broad involvement in numerous tissues and significant impact on both normal physiology and disease progression. Dysregulation of SPP1 has been strongly implicated in the pathogenesis and progression of several diseases, including cancer, cardiovascular diseases, autoimmune disorders, and chronic inflammatory conditions. The expression of SPP1 is tightly regulated by genetic and nongenetic mechanisms. Genetic alterations, such as single-nucleotide polymorphisms, insertions and deletions, and structural variations within the SPP1 gene, have been associated with increased susceptibility to various diseases, influencing disease severity and outcomes. Additionally, nongenetic regulations, including DNA methylation, histone modifications, and long noncoding RNAs, play crucial roles in modulating SPP1 expression in response to environmental and cellular signals. This review provides a comprehensive overview of the genetic and nongenetic regulatory mechanisms governing SPP1 and examines their implications in disease pathogenesis. By integrating recent findings, this review highlights the complex interplay between genetic predispositions and nongenetic regulations in determining SPP1 activity and offers new insights into its role as a potential biomarker and therapeutic target. Understanding these regulatory pathways is essential for the development of targeted interventions for diseases in which SPP1 plays a pivotal role.
Collapse
Affiliation(s)
- Sungju Jung
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jiseon Ha
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong Hoon Park
- Molecular Medicine Laboratory, Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Kyung Hyun Yoo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Republic of Korea; Research Institute of Women's Health, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| |
Collapse
|
2
|
Zhang S, Wang H, Meng Q, Lee WYW, Li Z, Sun S. Recent advances in osteonecrosis of the femoral head: a focus on mesenchymal stem cells and adipocytes. J Transl Med 2025; 23:592. [PMID: 40426076 DOI: 10.1186/s12967-025-06564-6] [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: 10/13/2024] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating orthopedic disease characterized by femoral head collapse and destruction of bone and articular cartilage, resulting in severe joint pain and loss of hip mobility. Bone marrow mesenchymal stem cells (BMSCs) exhibit multilineage differentiation potential, including osteoblasts, adipocytes, fibroblasts, chondrocytes, and neurocytes. The imbalance between osteogenesis and adipogenesis in BMSCs plays a critical role in ONFH pathogenesis. Factors such as alcohol consumption and glucocorticoid exposure promote adipogenic differentiation while inhibiting osteogenic differentiation, leading to excessive adipocyte accumulation, reduced bone formation, and vascular impairment. We highlight the molecular mechanisms underlying ONFH with a particular focus on the role of BMSCs and further discuss the involvement of adipocytes. Moreover, we suggest that the use of adipose-derived mesenchymal stem cells (ADMSCs) is a viable approach for stem cell therapy and may have immense potential in ONFH. Several signaling pathways, including the Wnt, TGFβ/BMP, and PI3K/AKT pathways, along with various RNAs and other regulators, govern the osteogenesis and adipogenesis of BMSCs. These signaling pathways target essential transcription factors, such as Runx2 for osteogenesis and PPARγ and C/EBPs for adipogenesis. Adipocytes and their secreted adipokines, including leptin and adiponectin, strongly influence ONFH progression. Emerging therapies involving ADMSCs show potential for promoting bone regeneration and neovascularization. Our review provides a comprehensive overview of the current understanding of ONFH mechanisms by focusing on mesenchymal stem cells and adipocytes and suggests future research directions for therapeutic interventions.
Collapse
Affiliation(s)
- Shilei Zhang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Haojue Wang
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Qi Meng
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250012, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| |
Collapse
|
3
|
Luo F, Yang Y, Li D, Mao R, Huang Y, Lu J, Zhu X, Wang K, Fan Y, Zhang X. Low-temperature plasma effect-induced enhancement of osteogenic activity in calcium phosphate ceramics. Acta Biomater 2025:S1742-7061(25)00301-0. [PMID: 40319990 DOI: 10.1016/j.actbio.2025.04.048] [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: 10/11/2024] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Calcium phosphate (Ca-P) ceramics are promising bioactive material that can be used for the remodeling and regeneration of bone tissue. However, it's sintering temperature-dependent mechanical strength, which is negatively correlated with its bioactivity, causes difficulties in improving the comprehensive performance of Ca-P ceramics. Here, the femtosecond laser (FSL) with low-temperature plasma effect was adopted to modify the hydroxyapatite (HA) ceramics after high temperatures (1250 °C) sintering, pursuing higher mechanical strength along with better osteogenic activity. The changes in the physicochemical properties of the materials and the osteogenic activity were characterized and investigated. Cell evaluations and in vivo experiments were performed to assess and verify the effect of FSL processing on the osteogenic capability of HA ceramics. The results indicated that α- and β-tricalcium phosphate (TCP) multiphase components were formed on the HA ceramic surfaces after laser treatment, simultaneously bringing about surface micro-nano porous structure, accelerated release of calcium (Ca) and phosphate (Pi) ions, enhancement of roughness, hydrophilicity and surface energy. Their synergistic effect facilitated apatite precipitation on the HA surface, promoted osteogenic differentiation and osteogenic/angiogenic gene expression. In vivo results also confirmed the enhancement of HA ceramic osteogenic activity by FSL treatment. This study presents an effective strategy of introducing FSL etching to high-temperature sintered Ca-P ceramics to improve the bone regeneration of HA ceramics and attain satisfactory mechanical strength at the same time. It will further promote the clinical application of HA ceramics in the field of bone regenerative repair. STATEMENT OF SIGNIFICANCE: This study introduces a method that uses the low-temperature plasma effect of the femtosecond laser (FSL) to modify the surfaces of high-temperature sintered hydroxyapatite (HA) ceramics, enhancing their osteogenic activity while maintaining the original mechanical strength. FSL processing induces the formation of bioactive multiphase of tricalcium phosphate (α-TCP and β-TCP) on the surfaces, creates micro-nano topographies, improves hydrophilicity and surface energy, promoting osteoblast differentiation and osteogenic gene expression for faster bone regeneration. This method overcomes the issue that high-temperature sintered HA ceramics have high strength but low osteogenic activity. It provides a modification method for HA ceramics with well-characterized performance enhancements, offering a convenient and effective strategy for high quality bone regenerative repair.
Collapse
Affiliation(s)
- Fengxiong Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yu Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Dongxuan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ruiqi Mao
- College of Materials Science and Engineering, Sichuan University, Chengdu 610064, China
| | - Yawen Huang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jian Lu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Research Center for Material Genome Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Research Center for Material Genome Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Chengdu 610064, China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Research Center for Material Genome Engineering, Sichuan University, Chengdu 610064, China; Provincial Engineering Research Center for Biomaterials Genome of Sichuan, Chengdu 610064, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Research Center for Material Genome Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
4
|
Brown MR, Soto-Feliciano YM. Menin: from molecular insights to clinical impact. Epigenomics 2025; 17:489-505. [PMID: 40152985 PMCID: PMC12026131 DOI: 10.1080/17501911.2025.2485019] [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: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 03/30/2025] Open
Abstract
Menin, the protein product of the MEN1 gene, is essential for development and has been implicated in multiple different cancer types. These include leukemias and several different solid tumors, including neuroendocrine tumors. Menin interacts with many different protein partners and genomic loci in a context-dependent manner, implicating it in numerous cellular processes. The role of Menin varies across tumor types as well, acting as a tumor suppressor in some tissues and an oncogenic co-factor in others. Given the role of Menin in cancer, and particularly its oncogenic role in acute myeloid leukemia, the development of Menin inhibitors has been an expanding field over the past 10-15 years. Many inhibitors have been in clinical trials and one has recently received approval from the Food and Drug Administration (FDA). In this review, we explore the role of Menin in multiple cancer types, the development of Menin inhibitors and their clinical applications and what the focus of the field should be in the next 5-10 years to expand the use and efficacy of these drugs.
Collapse
Affiliation(s)
- Margaret R. Brown
- Department is Biology, Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yadira M. Soto-Feliciano
- Department is Biology, Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
Zeng D, Yu J, Lu K, Yi D, Xia Z, Qin L, Xiao G, Yang X, Tong L, Chen D. Runx2 controls the osteogenic fate of growth plate chondrocytes. Genes Dis 2025; 12:101453. [PMID: 39872471 PMCID: PMC11761896 DOI: 10.1016/j.gendis.2024.101453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/09/2024] [Accepted: 11/02/2024] [Indexed: 01/30/2025] Open
Affiliation(s)
- Daofu Zeng
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Jiamin Yu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Ke Lu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Dan Yi
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Zhidao Xia
- Institute of Life Science, Swansea University Medical School, Swansea, SA2 8PP, UK
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 518172, China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing 100071, China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
6
|
Moghbeli M. MicroRNAs as the critical regulators of bone metastasis during prostate tumor progression. Int J Biol Macromol 2025; 309:142912. [PMID: 40203904 DOI: 10.1016/j.ijbiomac.2025.142912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men globally. Although, there are various therapeutic methods for the localized or advanced cancers, there is still a high rate of mortality among PCa patients that is mainly associated with bone metastasis in advanced tumors. There are few options available for treating bone metastasis in PCa, which only provide symptom relief without curing the disease. Therefore, it is crucial to evaluate the molecular mechanisms associated with bone metastasis of PCa cells to suggest the novel diagnostic and therapeutic approaches that could lower the morbidity and mortality rates in PCa patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological processes such as tumor growth and osteoblasts/osteoclasts formation. Since, miRNA deregulation has been also frequently observed in PCa patients with bone metastasis, we discussed the role of miRNAs in bone metastasis during PCa progression. It has been reported that miRNAs mainly reduced the ability of PCa tumor cells for the bone metastasis through the regulation of WNT, NF-kB, PI3K/AKT, and TGF-β signaling pathways. They also affected the EMT process, transcription factors, and structural proteins to regulate the bone metastasis during PCa progression. This review paves the way to suggest the miRNAs as the reliable markers not only for the non-invasive early diagnosis, but also for the targeted therapy of PCa tumors with bone metastasis.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Boisen IM, Krarup Knudsen N, Nielsen JE, Kooij I, Bagger ML, Kaludjerovic J, O'Shaughnessy P, Andrews PW, Ide N, Toft BG, Juul A, Mehmedbasic A, Jørgensen A, Smith LB, Norman R, Meyts ERD, Lanske B, Blomberg Jensen M. Changes in local mineral homeostasis facilitate the formation of benign and malignant testicular microcalcifications. eLife 2025; 13:RP95545. [PMID: 40279260 PMCID: PMC12029210 DOI: 10.7554/elife.95545] [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] [Indexed: 04/27/2025] Open
Abstract
Testicular microcalcifications consist of hydroxyapatite and have been associated with an increased risk of testicular germ cell tumors (TGCTs) but are also found in benign cases such as loss-of-function variants in the phosphate transporter SLC34A2. Here, we show that fibroblast growth factor 23 (FGF23), a regulator of phosphate homeostasis, is expressed in testicular germ cell neoplasia in situ (GCNIS), embryonal carcinoma (EC), and human embryonic stem cells. FGF23 is not glycosylated in TGCTs and therefore cleaved into a C-terminal fragment which competitively antagonizes full-length FGF23. Here, Fgf23 knockout mice presented with marked calcifications in the epididymis, spermatogenic arrest, and focally germ cells expressing the osteoblast marker Osteocalcin (gene name: Bglap, protein name). Moreover, the frequent testicular microcalcifications in mice with no functional androgen receptor and lack of circulating gonadotropins are associated with lower Slc34a2 and higher Bglap/Slc34a1 (protein name: NPT2a) expression compared with wild-type mice. In accordance, human testicular specimens with microcalcifications also have lower SLC34A2 and a subpopulation of germ cells express phosphate transporter NPT2a, Osteocalcin, and RUNX2 highlighting aberrant local phosphate handling and expression of bone-specific proteins. Mineral disturbance in vitro using calcium or phosphate treatment induced deposition of calcium phosphate in a spermatogonial cell line and this effect was fully rescued by the mineralization inhibitor pyrophosphate. In conclusion, testicular microcalcifications arise secondary to local alterations in mineral homeostasis, which in combination with impaired Sertoli cell function and reduced levels of mineralization inhibitors due to high alkaline phosphatase activity in GCNIS and TGCTs facilitate osteogenic-like differentiation of testicular cells and deposition of hydroxyapatite.
Collapse
Affiliation(s)
- Ida Marie Boisen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of CopenhagenCopenhagen NDenmark
| | - Nadia Krarup Knudsen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Ireen Kooij
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Mathilde Louise Bagger
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Jovanna Kaludjerovic
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | - Peter O'Shaughnessy
- School of Biodiversity, One Health & Veterinary Medicine, University of GlasgowGlasgowUnited Kingdom
| | - Peter W Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western BankSheffieldUnited Kingdom
| | - Noriko Ide
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | | | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Arnela Mehmedbasic
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Anne Jørgensen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research InstituteEdinburghUnited Kingdom
| | - Richard Norman
- Department of Urology, Dalhousie UniversityHalifaxCanada
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Beate Lanske
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | - Martin Blomberg Jensen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
- Department of Clinical Medicine, Copenhagen University HospitalCopenhagenDenmark
| |
Collapse
|
8
|
Saito A, Nagayama K, Okada H, Onodera S, Aida N, Nakamura T, Sawada T, Hojo H, Kato S, Azuma T. Downregulation of Nesprin1 by Runx2 deficiency is critical for the development of skeletal laminopathy-like pathology. Proc Natl Acad Sci U S A 2025; 122:e2320138122. [PMID: 40208950 PMCID: PMC12012476 DOI: 10.1073/pnas.2320138122] [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: 11/28/2023] [Accepted: 01/31/2025] [Indexed: 04/12/2025] Open
Abstract
Runx2 is a master regulator of bone formation, and its dysfunction causes cleidocranial dysplasia (CCD) in humans. When iPS cells were generated from patients with CCD and Runx2-deficient iPS cells were generated using gene-editing techniques, abnormal laminopathy-like nuclei were observed. Runx2-deficient cells showed reduced Lamin A/C expression, but not protein levels. However, in Runx2-deficient cells, both the gene expression and protein levels of Nesprin1 were reduced, perinuclear actin fibers were sparser, and nuclear stiffness was reduced. Forced expression of Lamin A/C increased nuclear stiffness but did not improve nuclear morphology. In contrast, the induction of Nesprin1 expression alone normalized nuclear stiffness and restored nuclear morphology and perinuclear actin distribution. In Runx2-null cells, mechanical stress-induced phosphorylation of emerin was not observed. In contrast, forced expression of Nesprin1 in Runx2-null cells resulted in phosphorylation of emerin, indicating the restoration of intracellular tension. These observations were confirmed by atomic force microscopy. Therefore, the intracellular tension was inferred to pull the nuclear membrane into its normal shape. CUT&RUN assay and single RNA-seq analysis showed that an aberrant nuclear membrane caused loss of nuclear lamina gene regulation machinery, making the progression of normal osteogenic differentiation impossible; however, supplementation with Nesprin1 restored gene regulation mechanisms and promoted preosteoblast formation with normal nuclear morphology. Nesprin1 expression induced by Runx2 is essential for epigenetic regulation of the nuclear lamina. We propose CCD as a type of laminopathy involving defective expression of Nesprin1 regulated by Runx2.
Collapse
Affiliation(s)
- Akiko Saito
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Kazuaki Nagayama
- Department of Mechanical Systems Engineering, Ibaraki University, Ibaraki316-8511, Japan
| | - Hiroyuki Okada
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Natsuko Aida
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
| | - Takashi Sawada
- Department of Histology and Developmental Biology, Tokyo Dental College, Tokyo101-0061, Japan
| | - Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| | - Shigeaki Kato
- Department of Pharmacology, Iryo Sosei University, Fukushima970-8551, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima973-8403, Japan
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, Tokyo101-0061, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo101-0061, Japan
- Obitsusankei Hospital, Saitama350-0021, Japan
| |
Collapse
|
9
|
Bergamasco MI, Ogier JM, Garnham AL, Whitehead L, Rogers K, Smyth GK, Burt RA, Voss AK, Thomas T. Loss of KAT6B causes premature ossification and promotes osteoblast differentiation during development. Dev Biol 2025; 520:141-154. [PMID: 39832706 DOI: 10.1016/j.ydbio.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
The MYST family histone acetyltransferase gene, KAT6B (MYST4, MORF, QKF) is mutated in two distinct human congenital disorders characterised by intellectual disability, facial dysmorphogenesis and skeletal abnormalities; the Say-Barber-Biesecker-Young-Simpson variant of Ohdo syndrome and Genitopatellar syndrome. Despite its requirement in normal skeletal development, the cellular and transcriptional effects of KAT6B in skeletogenesis have not been thoroughly studied. Here, we show that germline deletion of the Kat6b gene in mice causes premature ossification in vivo, resulting in shortened craniofacial elements and increased bone density, as well as shortened tibias with an expanded pre-hypertrophic layer, as compared to wild type controls. Mechanistically, we show that the loss of KAT6B in mesenchymal progenitor cells promotes transition towards an osteoblast-progenitor state with upregulation of gene targets of RUNX2, a master regulator of osteoblast development and concomitant downregulation of SOX9, a critical gene in chondrocyte development. Moreover, we find that compound heterozygosity at Kat6b and Runx2 loci partially rescues the reduction in ossification of Runx2 heterozygous, but not homozygous mice, suggesting that KAT6B may limit the action of RUNX2, possibly through a role in maintaining progenitors in an undifferentiated state. Moreover, our results show that KAT6B has essential roles in regulating the expression of a large number of genes involved in skeletogenesis and bone development.
Collapse
Affiliation(s)
- Maria I Bergamasco
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Jacqueline M Ogier
- The Department of Audiology and Speech Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Kelly Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rachel A Burt
- Department of Genetics, The Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, 3052, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
10
|
Lai YC, Yao ZK, Chang TC, Feng CW, Kuo TJ, Luo YW, Jean YH, Lin HYH, Wen ZH. Dextromethorphan Inhibits Osteoblast Differentiation and Bone Regeneration of Rats With Subcritical-Sized Calvarial Defects. ENVIRONMENTAL TOXICOLOGY 2025; 40:650-663. [PMID: 39607004 DOI: 10.1002/tox.24447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
The glutamatergic signaling pathway, which is mediated by N-methyl-D-aspartate (NMDA) receptors, is crucial for osteoblast differentiation and bone function. Dextromethorphan (DXM), a widely used antitussive, is a noncompetitive antagonist of the NMDA receptor. However, the effects of DXM on osteoblast and bone regeneration remain unclear. The present study investigated the effects of DXM on osteogenesis in vitro and in vivo. A MC3T3-E1 preosteoblast cell line was treated with varying concentrations of DXM. Real-time-quantitative polymerase chain reaction (RT-qPCR) and Western-blot analysis were performed to evaluate the expression of osteogenesis-related runt-related transcription factor 2 (RUNX2), osterix (OSX), osteocalcin (OCN), collagen type 1α (Col-1α), and alkaline phosphatase (ALP) after DXM treatment. Zebrafish embryos were incubated with DXM, which had potential to affect the ossification of the vertebrae and skull, and analyzed using calcein staining. Furthermore, we used a rat calvarial defect model to assess the effects of DXM on bone regeneration by using microcomputed tomography. The results indicate that DXM inhibited extracellular mineralization, ALP activity, and the expression of osteogenic markers, namely RUNX2, OSX, OCN, Col-1α, and ALP, in MC3T3-E1 cells. DXM suppressed skeleton ossification in zebrafish and affected bone regeneration in rats with calvarial defects. However, the mineral density of the regenerated bones did not differ significantly between the DXM and control groups. The present study demonstrated that DXM negatively affects the osteogenic function of osteoblasts, leading to impaired skeletal development and bone regeneration. Thus, clinicians should consider the negative effects of DXM on bone regeneration.
Collapse
Affiliation(s)
- Yu-Cheng Lai
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Orthopedics, Asia University Hospital, Taichung, Taiwan
| | - Zhi-Kang Yao
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Tien-Chieh Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chien-Wei Feng
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Jen Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Eternal Dental Clinic, Taichung, Taiwan
| | - Yi-Wei Luo
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Hugo Y-H Lin
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
- National Museum of Marine Biology & Aquarium, Pingtung, Taiwan
| |
Collapse
|
11
|
Bhuyan S, Swain S, Rautray TR. Polarised hydroxyapatite- sodium alginate composite as an antibacterial filler matrix. J Biol Phys 2025; 51:13. [PMID: 40095225 PMCID: PMC11914544 DOI: 10.1007/s10867-025-09679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Bone-substituted composite material based on bioceramics and polymer has enhanced their biological performance with dynamic properties such as bioactivity, biocompatibility, osseointegration, and mechanical stability, which can be used in a controlled drug delivery system for avoiding infections as well as pain. Here in this study, we developed a new approach for inducing antibacterial and osteogenic responses on biomaterial substrates via surface polarisation. The hydroxyapatite- sodium alginate composite was negatively polarised using a corona poling setup and characterised using X-ray diffraction analysis. The thermally stimulated depolarization current study showed a maximum current of 4.74 nA/cm2, observed at a temperature of 480 °C. The wettability of the specimen was measured using contact angle measurements, which demonstrated that the polarised composite specimen exhibited higher water retention ability. The bacterial cell viability test was measured using the 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay, which revealed poor bacterial growth on polarised specimens as compared to their unpolarised counterparts. In addition, the osteogenic MG63 cell proliferation showed increased gene expression on polarised specimens. These findings showed that polarising hydroxyapatite- sodium alginate composite could be an excellent option to be used as an antibacterial bone filler matrix for faster healing as it showed both antibacterial and osteogenic activity.
Collapse
Affiliation(s)
- Samapika Bhuyan
- Biomaterials and Tissue Regeneration Lab, Centre of Excellence, Siksha 'O'Anusandhan (Deemed to Be University), Odisha, Bhubaneswar, 751030, India
| | - Subhasmita Swain
- Biomaterials and Tissue Regeneration Lab, Centre of Excellence, Siksha 'O'Anusandhan (Deemed to Be University), Odisha, Bhubaneswar, 751030, India
| | - Tapash Ranjan Rautray
- Biomaterials and Tissue Regeneration Lab, Centre of Excellence, Siksha 'O'Anusandhan (Deemed to Be University), Odisha, Bhubaneswar, 751030, India.
| |
Collapse
|
12
|
Hayashi-Suzuki M, Fukuda-Tatano S, Kishimoto-Ogata M, Ehara-Kawagishi M, Ohminami H, Masuda M, Taketani Y. Maternal excess dietary phosphate intake in the periconceptional period is a potential risk for mineral disorders in offspring mice. Sci Rep 2025; 15:8844. [PMID: 40087383 PMCID: PMC11909113 DOI: 10.1038/s41598-025-91717-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Growing consumption of processed foods may cause a greater risk of excessive dietary phosphate intake. The increased dietary phosphate intake as a food additive in the periconceptional period may affect the children's future health. Here, we investigated the effects of maternal excess dietary phosphate intake on offspring in C57BL/6J mice. Female mice were fed a control diet (CP, 0.8% phosphate) or a high-phosphate diet (HP, 1.5% phosphate) for either 21 days during pre-pregnancy or almost 20 days during pregnancy. After weaning, offspring were raised on the CP diet. Relative to the CP groups, offspring from dams fed HP during pre-pregnancy or pregnancy showed decreased urinary phosphate excretion without significant changes in either plasma phosphate level or renal sodium-dependent phosphate transporter mRNA expression at 3 or 10 weeks. However, mRNA expression of intestinal sodium-dependent phosphate transporter was decreased, suggesting that the reduced urinary phosphate excretion was due to decreased absorption of intestinal phosphate. Interestingly, offspring in the HP groups also demonstrated significant differences in plasma levels of parathyroid hormone, fibroblast growth factor 23, and vitamin D. To our knowledge, this is the first report to show that maternal excess intake of dietary phosphate in the periconceptional period disturbs phosphate metabolism in offspring.
Collapse
Affiliation(s)
- Mayu Hayashi-Suzuki
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Shiori Fukuda-Tatano
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
- Department of Health and Nutrition, Faculty of Nursing and Nutrition, The University of Shimane, Izumo, Japan
| | - Maki Kishimoto-Ogata
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Miyu Ehara-Kawagishi
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Graduate School of Medical Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| |
Collapse
|
13
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
14
|
Miyasaka N, Torii D, Satomi T, Sakurai K, Nakahara T, W Tsutsui T. Aspirin promotes odontogenic differentiation via a mechanism involving FOXC1, RUNX2, and MCAM expression. J Oral Biosci 2025; 67:100622. [PMID: 39892782 DOI: 10.1016/j.job.2025.100622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES This study aimed to investigate the effects of aspirin on the early stages of odontogenic differentiation. The roles of FOXC1, RUNX2, and MCAM gene expression in the mechanism of odontogenic differentiation were evaluated by examining the effects of downregulated FOXC1 or RUNX2 expression using small interfering RNAs (siRNAs). METHODS Dental pulp cells were treated with aspirin (0, 2.5, 50, 100 μg/ml) to assess its impact on mineralization. The gene expression levels of FOXC1, RUNX2, and MCAM were measured using digital polymerase chain reaction, and the effects of siRNA-mediated knockdown of FOXC1 and RUNX2 were analyzed. The mineralization potential was quantitatively assessed using Alizarin Red S staining and a calcium assay. RESULTS Analysis of cell growth curves and doubling times indicated that aspirin did not affect cell proliferation at 2.5 μg/ml and 50 μg/ml; however, 50 μg/ml aspirin promoted mineralization. In the FOXC1 and RUNX2 knockdown experiments, fluctuations in FOXC1, RUNX2, and MCAM gene expression were observed in the aspirin-treated group, suggesting the involvement of these genes in mineralization. Alizarin red S staining and calcium assays further demonstrated that aspirin enhanced mineralization. CONCLUSIONS These findings indicate that aspirin promotes odontogenic differentiation and regulates the expression of FOXC1, RUNX2, and MCAM. This suggests that aspirin may serve as a promising new therapeutic agent in dental pulp regenerative medicine.
Collapse
Affiliation(s)
- Naoki Miyasaka
- Department of Oral and Maxillofacial Surgery, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Daisuke Torii
- Department of Pharmacology, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Takafumi Satomi
- Department of Oral and Maxillofacial Surgery, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Kenichi Sakurai
- Department of Surgery, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Takeo W Tsutsui
- Department of Pharmacology, The Nippon Dental University School of Life Dentistry at Tokyo, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| |
Collapse
|
15
|
Yi L, Han N, Li Z, Jiang H, Cao Z. Relaxin-2 promotes osteoblastic differentiation mediated by epidermal growth factor and epidermal growth factor receptor signaling. Biotechnol Appl Biochem 2025; 72:260-267. [PMID: 39219221 DOI: 10.1002/bab.2661] [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: 12/11/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Loss of osteogenic differentiation potential of osteoblasts has been associated with the pathogenesis of osteoporosis. Thus, stimulation of osteoblastic differentiation is a therapeutic strategy for osteoporosis. Relaxin-2 is a peptide hormone with potent biological functions. However, the effects of Relaxin-2 in osteoblastic differentiation and osteoporosis have not been reported before. Here, we report a novel physiological role of Relaxin-2 in promoting osteoblastic differentiation and mineralization of MC3T3-E1 cells. Our results indicate that exposure to Relaxin-2 upregulated the expression, and elevated the activity of alkaline phosphatase (ALP) when MC3T3-E1 cells were cultured in osteogenic differentiation medium (OM). Additionally, Relaxin-2 upregulated the mRNA levels of osteocalcin (ocn), osteopontin (opn), and collagen type I alpha 1 (Col1a1). The alizarin red S staining assay revealed that Relaxin-2 promoted the mineralization of MC3T3-E1 cells. We also found that Relaxin-2 increased the expression of Runx-2 as well as the epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR). Importantly, silencing of EGF abolished the effects of Relaxin-2 in osteoblastic differentiation and related gene expression. These findings suggest that Relaxin-2 stimulates osteogenic differentiation through activating EGF/EGFR signaling.
Collapse
Affiliation(s)
- Lankai Yi
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Ning Han
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhong Li
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Housen Jiang
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhenhao Cao
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| |
Collapse
|
16
|
Lee YH, Yi HK, Pradhan PM, Kim TK, Jang S. Effect of c-Myb overexpression on osteoblastic-, odontoblastic-, and cementoblastic differentiation of primary human periodontal ligament cells. Eur J Oral Sci 2025; 133:e13040. [PMID: 39865493 DOI: 10.1111/eos.13040] [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: 07/09/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
The periodontal ligament (PDL) is a connective tissue, and PDL cells have a potential to differentiate into cementoblasts, osteoblasts, and gingival fibroblasts. This study investigated whether transcription factor c-Myb could induce differentiation of PDL cells for periodontal regeneration. PDL cells were isolated from extracted teeth and cultured. c-Myb was transfected to PDL cells using replication-deficient adenoviral vector. Differentiation of the PDL cells was analyzed by immunoblot, alkaline phosphatase activity, Alizarin red stain, and immunofluorescence analysis. Cell viability on titanium surfaces was analyzed by crystal violet stain and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. PDL cells cultured in osteogenic medium showed increased production of osteogenic and cementogenic molecules. Moreover, c-Myb-transfected cells showed increased production of dentinogenic molecules, in addition to the osteogenic and cementogenic molecules, even in normal culture condition. c-Myb-transfected cells also exhibited increased autophagy and type I collagen production under nutrient deprivation. When grown on a titanium surface, c-Myb-transfected cells showed increased production of osteogenesis-, dentinogenesis-, and cementogenesis-related molecules and cell viability. Thus, these results suggest that c-Myb might play an essential role during periodontal regeneration by improving the differentiation of PDL cells, and c-Myb can be utilized for enhancing the attachment of PDL cells to dental implant surfaces.
Collapse
Affiliation(s)
- Young-Hee Lee
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si, South Korea
| | - Ho-Keun Yi
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si, South Korea
| | - Paras Man Pradhan
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si, South Korea
| | - Tae-Kun Kim
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si, South Korea
| | - Sungil Jang
- Department of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju-si, South Korea
| |
Collapse
|
17
|
Bao M, Wang X, Li X, Sun R, Wang Z, Jiang T, Wang H, Feng J. Single-Cell Landscape of the Cochlea Revealed Cell-Type-Specific Diversification in Hipposideros armiger Based on PacBio Long-Read Sequencing. Biomolecules 2025; 15:211. [PMID: 40001514 PMCID: PMC11853400 DOI: 10.3390/biom15020211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Echolocation represents one of the most rapid adaptive sensorimotor modulation behaviors observed in mammals, establishing bats as one of the most evolutionarily successful mammals. Bats rely on high-frequency hearing for survival, but our understanding of its cellular molecular basis is scattered and segmented. Herein, we constructed the first single-cell transcriptomic landscape of the cochlea in Hipposideros armiger, a CF-FM bat, using a PacBio-optimized genome and compared it with the results obtained from unoptimized original genomes. Sixteen distinct cell types were distributed across five spatial regions of the cochlea. Notably, through hematoxylin and eosin staining and fluorescence in situ hybridization, we identified new types of spiral ganglion neuron (SGN) cells in the cochlea of H. armiger. These SGN cells are likely critical for auditory perception and may have driven the adaptive evolution of high-frequency hearing in this species. Furthermore, we uncovered the differentiation relationships of among specific cell types, such as the transition from supporting cells to hair cells. Using the cochlear cell atlas as a reference, cell types susceptible to deafness-associated genes (in the human) were also identified. In summary, this study provides novel insights into the cellular and molecular mechanisms underlying the adaptive high-frequency hearing in bats and highlights potential candidate cell types and genes for therapeutic interventions in hearing loss.
Collapse
Affiliation(s)
- Mingyue Bao
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xue Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xintong Li
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Ruyi Sun
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Zhiqiang Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Hui Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| |
Collapse
|
18
|
Dong Q, Fu H, Li W, Ji X, Yin Y, Zhang Y, Zhu Y, Li G, Jia H, Zhang H, Wang H, Hu J, Wang G, Wu Z, Zhang Y, Xu S, Hou Z. Nuclear farnesoid X receptor protects against bone loss by driving osteoblast differentiation through stabilizing RUNX2. Bone Res 2025; 13:20. [PMID: 39885145 PMCID: PMC11782663 DOI: 10.1038/s41413-024-00394-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025] Open
Abstract
The delicate balance between bone formation by osteoblasts and bone resorption by osteoclasts maintains bone homeostasis. Nuclear receptors (NRs) are now understood to be crucial in bone physiology and pathology. However, the function of the Farnesoid X receptor (FXR), a member of the NR family, in regulating bone homeostasis remains incompletely understood. In this study, in vitro and in vivo models revealed delayed bone development and an osteoporosis phenotype in mice lacking FXR in bone marrow mesenchymal stem cells (BMSCs) and osteoblasts due to impaired osteoblast differentiation. Mechanistically, FXR could stabilize RUNX2 by inhibiting Thoc6-mediated ubiquitination, thereby promoting osteogenic activity in BMSCs. Moreover, activated FXR could directly bind to the Thoc6 promoter, suppressing its expression. The interaction between RUNX2 and Thoc6 was mediated by the Runt domain of RUNX2 and the WD repeat of Thoc6. Additionally, Obeticholic acid (OCA), an orally available FXR agonist, could ameliorate bone loss in an ovariectomy (OVX)-induced osteoporotic mouse model. Taken together, our findings suggest that FXR plays pivotal roles in osteoblast differentiation by regulating RUNX2 stability and that targeting FXR may be a promising therapeutic approach for osteoporosis.
Collapse
Affiliation(s)
- Qi Dong
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haoyuan Fu
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenxiao Li
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinyu Ji
- Department of Cardiology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yingchao Yin
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiran Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Yanbo Zhu
- Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Hebei Food Inspection and Research Institute, Shijiazhuang, Hebei, China
| | - Guoqiang Li
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huiyang Jia
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Heng Zhang
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haofei Wang
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinglue Hu
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | | | - Zhihao Wu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, Anhui, China
| | - Yingze Zhang
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sujuan Xu
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory for Diabetic Kidney Disease, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nephrology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Orthopaedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
19
|
Shen Q, Hu W, Liu F, Dong S, Peng X, Zhong Y, Chen C, Zuo Y, Ge C, Li W, Zha W, Ye Z, Cao Z, Liao L. Dipropyl phthalate induces craniofacial chondrogenic defects in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117603. [PMID: 39721426 DOI: 10.1016/j.ecoenv.2024.117603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Dipropyl phthalate (DPRP), a plasticizer commonly utilized in the plastics industry, has been identified in food and the environment and has the potential to present a hazard to human health and the environment. In this study, the first comprehensive evaluation of DPRP-induced craniofacial chondrogenic defects was conducted using a zebrafish model. Zebrafish embryos were exposed to 1, 2, and 4 mg/L DPRP from 6 to 96 h post-fertilization. At 80 hpf, it was observed that exposure to DPRP resulted in craniofacial developmental malformations, which were mainly characterized by the shortening of the mandibular pharyngeal arches and the inability of the accompanying artery to elongate forward. The resulting phenotype was similar to that of micrognathia syndrome. Transcriptome sequencing and molecular docking analyses revealed that DPRP down-regulated chondrocyte-related genes and induced activation of the FoxO signaling pathway, which in turn interfered with cell proliferation and apoptosis. In this process, DPRP induced elevated levels of oxidative stress in the craniofacial pharyngeal arch while promoting inflammatory responses. This ultimately led to craniofacial chondrogenic malformations in zebrafish. The present study demonstrates that DPRP induces developmental toxicity of zebrafish craniofacial cartilage, which may have adverse effects on other aquatic organisms and humans.
Collapse
Affiliation(s)
- Qinyuan Shen
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Oral Diseases, Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang, Jiangxi 330006, PR China
| | - Weitao Hu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Fasheng Liu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Si Dong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Xinya Peng
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Yihang Zhong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Chao Chen
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Yuhua Zuo
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Chenkai Ge
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Weirong Li
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Wenwen Zha
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Zhijun Ye
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China.
| | - Lan Liao
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Oral Diseases, Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang, Jiangxi 330006, PR China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs and Epigenetics, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, Jiangxi 343009, PR China; The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
20
|
Ozisin MS, Imren G, Aydin B, Karaosmanoglu B, Taskiran EZ. The effect of LARP7 on gene expression during osteogenesis. Mol Biol Rep 2025; 52:120. [PMID: 39804499 DOI: 10.1007/s11033-024-10216-1] [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: 06/26/2024] [Accepted: 12/30/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND La-related protein 7 (LARP7) is a key regulator of RNA metabolism and is thought to play a role in various cellular processes. LARP7 gene autosomal recessive mutations are the cause of Alazami syndrome, which presents with skeletal abnormalities, intellectual disabilities, and facial dysmorphisms. This study aimed to determine the role of LARP7 in modulating gene expression dynamics during osteogenesis. METHODS AND RESULTS First, the temporal expression profile of the LARP7 gene during various stages of osteogenesis was examined. Then, RNA interference-mediated knockdown of LARP7 was implemented and high-throughput RNA-seq analysis was performed in order to identify global gene expression changes associated with knockdown of LARP7. The findings show there were significant alterations in the overall gene expression profile. The observed down-regulation in extracellular matrix (ECM) component genes suggests that it might lead to impairments in the structure and function of the bone matrix. Additionally, modulation of alternative splicing events were observed, especially in the RUNX2 and SPP1, indicating the potential contribution of LARP7 to the phenotypic features observed in Alazami syndrome. CONCLUSION Overall, the findings clarify the regulatory mechanisms of LARP7 in osteogenic differentiation and illuminate potential avenues for therapeutic interventions in patients with skeletal disorders.
Collapse
Affiliation(s)
- M Samil Ozisin
- Institute of Health Sciences, Department of Medical and Surgical Research, Hacettepe University, Ankara, Turkey
| | - Gozde Imren
- Institute of Health Sciences, Department of Medical and Surgical Research, Hacettepe University, Ankara, Turkey
- Faculty of Medicine, Department of Medical Genetics, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Busra Aydin
- Institute of Health Sciences, Department of Medical and Surgical Research, Hacettepe University, Ankara, Turkey
| | - Beren Karaosmanoglu
- Faculty of Medicine, Department of Medical Genetics, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Ekim Z Taskiran
- Institute of Health Sciences, Department of Medical and Surgical Research, Hacettepe University, Ankara, Turkey.
- Faculty of Medicine, Department of Medical Genetics, Hacettepe University, Sihhiye, Ankara, Turkey.
| |
Collapse
|
21
|
Bathina S, Prado M, Fuenmayor Lopez V, Colleluori G, Aguirre L, Chen R, Villareal DT, Armamento-Villareal R. PRDM16 Enhances Osteoblastogenic RUNX2 via Canonical WNT10b/β-CATENIN Pathway in Testosterone-Treated Hypogonadal Men. Biomolecules 2025; 15:79. [PMID: 39858473 PMCID: PMC11764227 DOI: 10.3390/biom15010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
We previously reported that PRDM16 mediated the improvement in body composition in testosterone (T)-treated hypogonadal men by shifting adipogenesis to myogenesis. Previous preclinical studies suggest that Prdm16 regulates Runx2, an important osteoblastic transcription factor, expression and activity. However, the changes in PRDM16, and other genes/proteins involved in osteoblastogenesis with T therapy in hypogonadal men are unexplored. We investigated the role of PRDM16 in RUNX2 activation by measuring changes in gene expression in peripheral blood monocytes (PBMCs) and proteins in the serum of hypogonadal men after T therapy for 6 months. Likewise, we evaluated changes in the WNT10b-β-CATENIN signaling pathway by gene expression and protein analyses. We found significant increases in PRDM16 and RUNX2 expression in PBMCs together with significant increases in serum proteins at 6 months when compared to baseline. There were also increases in gene and protein expressions of WNT10b, and β-CATENIN at 6 months. Furthermore, we found a significant positive correlation between % changes in PRDM16 and WNT10b. Our results suggest that T therapy activates PRDM16, leading to enhanced signaling in the canonical WNT10b-β-CATENIN-RUNX2 pathway, the pathway involved in osteoblastogenesis. The above findings may account for the improvement in bone density and quality in hypogonadal men treated with T.
Collapse
Affiliation(s)
- Siresha Bathina
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Mia Prado
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Virginia Fuenmayor Lopez
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Georgia Colleluori
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lina Aguirre
- Department of Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87107, USA
- Department of Medicine, New Mexico VA Health Care System, Albuquerque, NM 87107, USA
| | - Rui Chen
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Dennis T. Villareal
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Reina Armamento-Villareal
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| |
Collapse
|
22
|
Hudacova E, Abaffy P, Kaplan MM, Krausova M, Kubista M, Machon O. Single-cell transcriptomic resolution of osteogenesis during craniofacial morphogenesis. Bone 2025; 190:117297. [PMID: 39461490 DOI: 10.1016/j.bone.2024.117297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Craniofacial morphogenesis depends on complex cell fate decisions during the differentiation of post-migratory cranial neural crest cells. Molecular mechanisms of cell differentiation of mesenchymal cells to developing bones, cartilage, teeth, tongue, and other craniofacial tissues are still poorly understood. We performed single-cell transcriptomic analysis of craniofacial mesenchymal cells derived from cranial NCCs in mouse embryo. Using FACS sorting of Wnt1-Cre2 progeny, we carefully mapped the cell heterogeneity in the craniofacial region during the initial stages of cartilage and bone formation. Transcriptomic data and in vivo validations identified molecular determinants of major cell populations involved in the development of lower and upper jaw, teeth, tongue, dermis, or periocular mesenchyme. Single-cell transcriptomic analysis of Meis2-deficient mice revealed critical gene expression differences, including increased osteogenic and cell adhesion markers. This leads to affected mesenchymal cell differentiation and increased ossification, resulting in impaired bone, cartilage, and tongue formation.
Collapse
Affiliation(s)
- Erika Hudacova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 12000 Prague, Czech Republic.
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Mehmet Mahsum Kaplan
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Michaela Krausova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Ondrej Machon
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| |
Collapse
|
23
|
Zeng D, Umar M, Zhu Z, Pan H, Lu WW, Xiao G, Chen Y, Tong L, Chen D. Development of novel osteoarthritis therapy by targeting AMPK-β-catenin-Runx2 signaling. Genes Dis 2025; 12:101247. [PMID: 39552787 PMCID: PMC11566674 DOI: 10.1016/j.gendis.2024.101247] [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: 09/25/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 11/19/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating chronic joint disease affecting large populations of patients, especially the elderly. The pathological mechanisms of OA are currently unknown. Multiple risk factors are involved in OA development. Among these risk factors, alterations of mechanical loading in the joint leading to changes in biological signaling pathways have been known as a key event in OA development. The importance of AMPK-β-catenin-Runx2 signaling in the initiation and progression of OA has been recognized in recent years. In this review, we discuss the recent progress in understanding the role of this signaling pathway and the underlying interaction mechanisms during OA development. We also discuss the drug development aiming to target this signaling pathway for OA treatment.
Collapse
Affiliation(s)
- Daofu Zeng
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Muhammad Umar
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Zhenglin Zhu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haobo Pan
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, Guangdong 518071, China
| | - William W. Lu
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yan Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
24
|
Buchanan C, Chen S, Yuan Y, Guo T, Feng J, Zhang M, Carey G, Howard I, Sanchez J, Ho TV, Chai Y. Loss of Runx2 in Gli1 + osteogenic progenitors prevents bone loss following ovariectomy. JBMR Plus 2025; 9:ziae141. [PMID: 39996169 PMCID: PMC11848843 DOI: 10.1093/jbmrpl/ziae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 02/26/2025] Open
Abstract
Osteoporosis is a metabolic bone disorder characterized by low bone mass and bone mineral density. It is the most prevalent bone disease and a common cause of fracture in aging adults. Low bone mass, as seen in osteoporosis, results from an imbalance between osteoblast and osteoclast activity. Gli1+ cells are indispensable to the maintenance of bone tissue homeostasis. These cells give rise to osteoprogenitors and are present at the osteogenic fronts of long bones in adult mice. Runx2 is a key regulator of osteogenesis and plays a crucial role in osteoblastic differentiation and maturation during development. However, its function in maintaining adult bone tissue homeostasis remains unclear. In this study, we investigated the role of Runx2 in maintaining adult bone homeostasis in the context of ovariectomy-induced estrogen deficiency, a model for postmenopausal osteoporosis. Our results show that deletion of Runx2 in the Gli1+ osteogenic progenitor population prevents loss of both cortical and trabecular bone mass and mineralization after ovariectomy. At the cellular level, loss of Runx2 leads to a decrease in osteoclast activity. Our study indicates that Runx2 is essential for maintaining adult bone tissue homeostasis by regulating osteoclast differentiation.
Collapse
Affiliation(s)
- Connor Buchanan
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Shuo Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Yuan Yuan
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Grace Carey
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Ishmael Howard
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Janet Sanchez
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
25
|
Komori T. Bone development by Hedgehog and Wnt signaling, Runx2, and Sp7. J Bone Miner Metab 2025; 43:33-38. [PMID: 39352550 DOI: 10.1007/s00774-024-01551-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/25/2024] [Indexed: 04/01/2025]
Abstract
Hedgehog and canonical Wnt signaling pathways and the transcription factors Runx2 and Sp7 are essential for osteoblast differentiation. Ihh is necessary for the commitment of perichondrial mesenchymal cells to Runx2+ osteoprogenitors and for the formation of the bone collar and primary spongiosa. Runx2 is needed for osteoblast differentiation during both endochondral and intramembranous ossification. It regulates the commitment of mesenchymal cells to osteoblast-lineage cells and their proliferation by inducing the expression of Hedgehog, Fgf, Wnt, Pthlh signaling pathway genes, and Dlx5. The Runx2-induced expression of Fgfr2 and Fgfr3 is important for the proliferation of osteoblast-lineage cells. Runx2 induces Sp7 expression and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Runx2, Sp7, and canonical Wnt signaling induce the differentiation of preosteoblasts into osteoblasts. Canonical Wnt signaling, but not Sp7, enhances the proliferation of osteoblast-lineage cells. In mature osteoblasts, Runx2 plays an important role in the expression of major bone matrix protein genes, including Col1a1, Col1a2, Spp1, Ibsp, and Bglap/Bglap2. The canonical Wnt signaling pathway is also crucial for bone formation by mature osteoblasts. Sp7 is needed for osteocytes to acquire a sufficient number of processes and a reduction in these processes results in osteocyte apoptosis and cortical porosity.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan.
| |
Collapse
|
26
|
Sory DR, Heyraud ACM, Jones JR, Rankin SM. Ionic release from bioactive SiO 2-CaO CME/poly(tetrahydrofuran)/poly(caprolactone) hybrids drives human-bone marrow stromal cell osteogenic differentiation. BIOMATERIALS ADVANCES 2025; 166:214019. [PMID: 39326252 DOI: 10.1016/j.bioadv.2024.214019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
This study demonstrates that dissolution products of inorganic/organic SiO2-CaOCME/PTHF/PCL-diCOOH hybrid (70S30CCME-CL) drive human bone marrow stromal cells (h-BMSCs) down an osteogenic pathway with the production of mineralised matrix. We investigated osteogenesis through combined analyses of mRNA dynamics for key markers and targeted staining of mineralised matrix. We demonstrate that h-BMSCs undergo accelerated differentiation in vitro in response to the 70S30CCME-CL ionic milieu, as compared to incubation with osteogenic media. Extracts from 70S30CCME-CL promote osteogenesis by inducing changes in cellular metabolic activity, promoting changes in cell morphology consistent with the osteogenic lineage, and by enhancing mineralisation of hydroxyapatite in the extracellular matrix. Additionally, our results show that 70S30CCME-CL hybrids prove sustained functional resilience by maintaining osteostimulatory effects despite cumulated dissolution cycles. In co-differentiation medium, 70S30CCME-CL ionic release can modulate signalling pathways associated with non-osteogenic functions, further supporting their potential for bone regeneration applications. Overall, our study provides compelling experimental evidence that the 70S30CCME-CL hybrid is a promising biomaterial for bone tissue regeneration applications.
Collapse
Affiliation(s)
- David R Sory
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | | - Julian R Jones
- Department of Materials, Imperial College London, London, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
27
|
Hengtrakul N, Furrow E, Borofsky M, Toth F, Lulich JP. Expression of osteogenic proteins in kidneys of cats with nephrocalcinosis. J Vet Intern Med 2025; 39:e17278. [PMID: 39757788 PMCID: PMC11702495 DOI: 10.1111/jvim.17278] [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: 05/26/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Nephrocalcinosis is a common pathological finding in cats with chronic kidney disease and nephrolithiasis. Understanding its pathogenesis may identify future therapeutic targets. HYPOTHESIS Nephrocalcinosis is associated with expression of an osteogenic phenotype. ANIMALS Kidneys with medullary mineralization were obtained from 18 cats (10 with and 8 without nephroliths) undergoing necropsy. METHODS Cross-sectional study. Microradiography and histopathology (modified von Kossa stain) were used to confirm parenchymal mineralization. Immunohistochemistry for 5 osteogenic markers was performed to determine their co-localization with nephrocalcinosis. The proportion of kidneys with stronger immunointensity in mineralized versus non-mineralized regions was analyzed using 1-tailed sign tests. The proportion of kidneys with co-localization of nephrocalcinosis and each marker was compared between kidneys with and without nephroliths using Fisher's exact tests. RESULTS Nephrocalcinosis co-localized with osteopontin immunoreactivity in all 18 cats (100%) and with osteocalcin in 12 cats (67%). Both osteogenic markers had stronger immunointensity in mineralized regions compared with non-mineralized regions. Limited co-localization was observed with other markers: bone morphogenic protein-2 in 2 kidneys (both with nephroliths) and tissue non-specific alkaline phosphatase in 1 kidney (without nephroliths); runt-related transcription factor-2 was undetected. No statistically significant differences were found in the co-localization of nephrocalcinosis with osteogenic proteins between kidneys with and without nephroliths. CONCLUSIONS AND CLINICAL IMPORTANCE Expression of osteogenic proteins in areas of nephrocalcinosis indicates that nephrocalcinosis is associated with the development of an osteogenic phenotype. Targeting these processes could offer a novel approach to prevent nephrolithiasis at its origin.
Collapse
Affiliation(s)
- Nuttha Hengtrakul
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Eva Furrow
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Michael Borofsky
- Department of Urology, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ferenc Toth
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Jody P. Lulich
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| |
Collapse
|
28
|
Hojo H, Tani S, Ohba S. Modeling of skeletal development and diseases using human pluripotent stem cells. J Bone Miner Res 2024; 40:5-19. [PMID: 39498496 DOI: 10.1093/jbmr/zjae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/28/2024] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
Human skeletal elements are formed from distinct origins at distinct positions of the embryo. For example, the neural crest produces the facial bones, the paraxial mesoderm produces the axial skeleton, and the lateral plate mesoderm produces the appendicular skeleton. During skeletal development, different combinations of signaling pathways are coordinated from distinct origins during the sequential developmental stages. Models for human skeletal development have been established using human pluripotent stem cells (hPSCs) and by exploiting our understanding of skeletal development. Stepwise protocols for generating skeletal cells from different origins have been designed to mimic developmental trails. Recently, organoid methods have allowed the multicellular organization of skeletal cell types to recapitulate complicated skeletal development and metabolism. Similarly, several genetic diseases of the skeleton have been modeled using patient-derived induced pluripotent stem cells and genome-editing technologies. Model-based drug screening is a powerful tool for identifying drug candidates. This review briefly summarizes our current understanding of the embryonic development of skeletal tissues and introduces the current state-of-the-art hPSC methods for recapitulating skeletal development, metabolism, and diseases. We also discuss the current limitations and future perspectives for applications of the hPSC-based modeling system in precision medicine in this research field.
Collapse
Affiliation(s)
- Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoichiro Tani
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
29
|
Fang T, Zhang R, Song F, Chu X, Fu Q, Wu Q. miR-468-3p suppresses osteogenic differentiation of BMSCs by targeting Runx2 and inhibits bone formation. J Orthop Surg Res 2024; 19:887. [PMID: 39734217 DOI: 10.1186/s13018-024-05410-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
An improved understanding of the molecular actions underpinning bone marrow mesenchymal stem cell (BMSC) differentiation could highlight new therapeutics for osteoporosis (OP). Current evidence indicates that microRNAs (miRNAs) exert critical roles in many biological systems, including osteoblast differentiation. In this study, we examined miR-468-3p effects on osteogenic differentiation (OD). Distinct miR-468-3p reductions were identified during OD. MiR-468-3p also suppressed BMSC OD in gain- and loss-of-function assays, while it negatively regulated Runx2 as shown by molecular, protein, and bioinformatics approaches. When Runx2 was inhibited by small-interfering RNA (siRNA), the inhibitory effects of miR-468-3p toward BMSC osteogenesis were considerably reversed. Also, silenced miR-468-3p in ovariectomized (OVX) and sham mice augmented bone mass (BM) and bone formation (BF) and improved trabecular (Tb) microarchitecture. Therefore, miR-468-3p is a novel Runx2 regulator with key physiological action in BF and OD.
Collapse
Affiliation(s)
- Tao Fang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Ranxi Zhang
- Department of Spine Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Feng Song
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Xueru Chu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, 266000, China
| | - Qin Fu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Qianqian Wu
- Department of Cardiology, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong, 266000, China.
| |
Collapse
|
30
|
Wang W, Sun DF, Dong Z, Zhang WL. Icariin suppresses osteogenic differentiation and promotes bone regeneration in Porphyromonas gingivalis-infected conditions through EphA2-RhoA signaling pathway. Int Immunopharmacol 2024; 143:113302. [PMID: 39388889 DOI: 10.1016/j.intimp.2024.113302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/14/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Periodontitis is associated with multiple systemic diseases and can cause bone loss. Porphyromonas gingivalis (P. gingivalis) is one of the most virulent periodontal pathogens. Icariin is a flavonoid extracted from the traditional Chinese herbal medicine Herba Epimedii, and can regulate bone metabolism. However, its effects on promoting bone metabolism have not been fully elucidated. In this experiment, we infected MC3T3-E1 cells with P. gingivalis. Flow cytometry results show that persistent bacterial infection does not affect cell proliferative activity. Western blotting, ALP activity detection, mineral content determination, and immunofluorescence blotting confirmed that icariin improved osteogenic differentiation in the inflammatory state, and this effect may be more obvious in the early stage of osteogenic differentiation. The antibacterial assays, ROS and MMP fluorescence assays demonstrated that icariin exerted a significant inhibitory effect on bacterial growth and attenuated the inflammatory response in bacterial-infected conditions. The results of in vivo experiments in animals further validated the excellent properties exerted by icariin in the repair of bone defects. Additionally, in the P. gingivalis-infected state, icariin exert a regulatory effect on EphA2-RhoA signaling pathway to augment osteogenic differentiation. These exciting findings suggest that icariin holds significant potential for therapeutic application in the management of periodontal bone loss.
Collapse
Affiliation(s)
- Wei Wang
- Jinzhou Medical University, Jinzhou 121000, China
| | - Dan-Fang Sun
- Jinzhou Medical University, Jinzhou 121000, China
| | - Zhe Dong
- Jinzhou Medical University, Jinzhou 121000, China
| | - Wen-Lu Zhang
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
31
|
Alexander KA, Tseng HW, Lao HW, Girard D, Barbier V, Ungerer JPJ, McWhinney BC, Samuel SG, Fleming W, Winkler IG, Salga M, Genêt F, Banzet S, Ruitenberg MJ, Lévesque JP. A glucocorticoid spike derails muscle repair to heterotopic ossification after spinal cord injury. Cell Rep Med 2024; 5:101849. [PMID: 39657663 PMCID: PMC11722129 DOI: 10.1016/j.xcrm.2024.101849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 08/02/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Why severe injury to the central nervous system (CNS) triggers the development of large neurogenic heterotopic ossifications (NHOs) within periarticular muscles remains unknown. We report that spinal cord injury (SCI) triggers a rapid corticosterone spike in mice, which is causal for NHO development because treatments with corticosterone or the synthetic glucocorticoid (GC) receptor (GR) agonist dexamethasone are sufficient to trigger heterotopic ossification and upregulate the expression of osteoinductive and osteogenic differentiation genes in injured muscles even without SCI. The central role for GR signaling in causing NHO is further demonstrated in mice deleted for the GR gene (Nr3c1), which no longer develop NHO after SCI. Furthermore, administration of clinical GR antagonists inhibits NHO development in mice with SCI. This study identifies endogenous GC as causing pathological NHO after CNS injury and suggests that GR antagonists may be of prophylactic use to prevent NHO development in victims of severe CNS injuries.
Collapse
Affiliation(s)
- Kylie A Alexander
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Hsu-Wen Tseng
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Hong Wa Lao
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Dorothée Girard
- Institut de Recherche Biomédicale des Armées, 92140 Clamart, France; INSERM, UMR-MD U1197 SToRM, 92140 Clamart, France
| | - Valérie Barbier
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Jacobus P J Ungerer
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia; Department of Chemical Pathology, Pathology Queensland, Herston, QLD 4029, Australia
| | - Brett C McWhinney
- Department of Chemical Pathology, Pathology Queensland, Herston, QLD 4029, Australia
| | - Selwin G Samuel
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Whitney Fleming
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Ingrid G Winkler
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Marjorie Salga
- Unité Péri-Opératoire du Handicap, Physical and Rehabilitation Medicine Department, Hôpital Raymond-Poincaré, Assistance Publique Hôpitaux de Paris (APHP), 92380 Garches, France
| | - François Genêt
- Unité Péri-Opératoire du Handicap, Physical and Rehabilitation Medicine Department, Hôpital Raymond-Poincaré, Assistance Publique Hôpitaux de Paris (APHP), 92380 Garches, France; Université Versailles Saint-Quentin-en-Yvelines, UFR Simone Veil - Santé, END:ICAP, INSERM U1179, 78180 Montigny-le-Bretonneux, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées, 92140 Clamart, France; INSERM, UMR-MD U1197 SToRM, 92140 Clamart, France
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Jean-Pierre Lévesque
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
32
|
Jia X, Zhang G, Yu D. Application of extracellular vesicles in diabetic osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1466775. [PMID: 39720256 PMCID: PMC11666354 DOI: 10.3389/fendo.2024.1466775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/15/2024] [Indexed: 12/26/2024] Open
Abstract
As the population ages, the occurrence of osteoporosis is becoming more common. Diabetes mellitus is one of the factors in the development of osteoporosis. Compared with the general population, the incidence of osteoporosis is significantly higher in diabetic patients. Diabetic osteoporosis (DOP) is a metabolic bone disease characterized by abnormal bone tissue structure due to hyperglycemia and insulin resistance, reduced bone strength and increased risk of fractures. This is a complex mechanism that occurs at the cellular level due to factors such as blood vessels, inflammation, and hyperglycemia and insulin resistance. Although the application of some drugs in clinical practice can reduce the occurrence of DOP, the incidence of fractures caused by DOP is still very high. Extracellular vesicles (EVs) are a new communication mode between cells, which can transfer miRNAs and proteins from mother cells to target cells through membrane fusion, thereby regulating the function of target cells. In recent years, the role of EVs in the pathogenesis of DOP has been widely demonstrated. In this article, we first describe the changes in the bone microenvironment of osteoporosis. Second, we describe the pathogenesis of DOP. Finally, we summarize the research progress and challenges of EVs in DOP.
Collapse
Affiliation(s)
- Xiaopeng Jia
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Gongzi Zhang
- Department of Rehabilitation Medicine, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Deshui Yu
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
33
|
Lu ZJ, Gu HY, Li ZQ, Lin FX. Low frequency‑pulsed electromagnetic fields promote osteogenic differentiation of bone marrow‑derived mesenchymal stem cells by regulating connexin 43 expression. Exp Ther Med 2024; 28:446. [PMID: 39386938 PMCID: PMC11462399 DOI: 10.3892/etm.2024.12736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 08/22/2024] [Indexed: 10/12/2024] Open
Abstract
The present study investigated the effect of connexin 43 (Cx43) on the regulation of osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (BMSCs) using low-frequency-pulsed electromagnetic fields (LPEMF). The BMSCs were isolated and cultured in vitro using adherent whole-bone marrow cultures. CCK-8 assay was used to detect the effects of LPEMF on the proliferation ability of BMSCs and alkaline phosphatase (ALP) activity and the levels of osteogenic marker genes were detected to evaluate the osteogenic ability change following LPEMF treatment. Lentiviral vector-mediated RNA interference was transfected into BMSCs to inhibit the expression of Cx43 and western blotting was used to detect Cx43 expression. The BMSCs showed the highest proliferation following LPEMF treatment at 80 Hz for 1 h. The results of ALP activity, osteogenic marker genes and Alizarin Red S staining showed that the osteogenic ability was notably increased following LPEMF treatment at 80 Hz for 1 h. Cx43 expression increased during the osteogenic differentiation of BMSCs following LPEMF treatment at 80 Hz. The enhanced osteogenic differentiation of the LPEMF-treated BMSCs were partially reversed when Cx43 expression was inhibited. LPEMF may promote the osteogenic differentiation of BMSCs by regulating Cx43 expression and enhancing osteogenic ability.
Collapse
Affiliation(s)
- Zhi-Jun Lu
- Department of Spine Surgery, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Jiangxi Medical College of Nanchang University, Ganzhou Hospital-Nanfang Hospital of Southern Medical University), Ganzhou, Jiangxi 341000, P.R. China
| | - Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Jiangxi Medical College of Nanchang University, Ganzhou Hospital-Nanfang Hospital of Southern Medical University), Ganzhou, Jiangxi 341000, P.R. China
| | - Zhi-Qiang Li
- Department of Spine Surgery, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Jiangxi Medical College of Nanchang University, Ganzhou Hospital-Nanfang Hospital of Southern Medical University), Ganzhou, Jiangxi 341000, P.R. China
| | - Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Jiangxi Medical College of Nanchang University, Ganzhou Hospital-Nanfang Hospital of Southern Medical University), Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
34
|
Song J, Zhang Y, Jin X, Zhu Y, Li Y, Hu M. Eucommia ulmoides Oliver polysaccharide alleviates glucocorticoid-induced osteoporosis by stimulating bone formation via ERK/BMP-2/SMAD signaling. Sci Rep 2024; 14:29647. [PMID: 39609585 PMCID: PMC11604974 DOI: 10.1038/s41598-024-80859-4] [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: 07/24/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Osteoporosis (OP) is a metabolic disease characterized by low bone mineral mass owing to osteoblast dysfunction. Eucommia ulmoides Oliver (EuO) is a Chinese herbal medicine traditionally used to treat OP. Here, a polysaccharide purified from the EuO cortex (EuOCP3) was administered to OP mice constructed with dexamethasone (Dex) to investigate its anti-OP activity. EuOCP3 significantly improved Dex-induced bone microarchitecture destruction, increased osteoblast numbers and surface, and stimulated an increase in the expression of osteoblast differentiation markers in the femurs of OP mice. Furthermore, EuOCP3 was applied to MC3T3-E1 cells to further explore its effects on osteoblast differentiation. EuOCP3 significantly promoted osteoblast differentiation and increased the level of phosphorylated extracellular signal-regulated kinase1/2 (ERK1/2) and SMAD1/5/8. The EuOCP3-mediated enhancement of osteoblast differentiation-related proteins and phosphorylated SMAD1/5/8 expression levels was strongly suppressed by an ERK inhibitor (PD98059), which confirmed the critical role of ERK signaling in EuOCP3-induced osteoblast differentiation. In summary, EuOCP3 can stimulate bone formation by improving osteoblast differentiation via ERK/BMP-2/SMAD signaling, indicating the potential use of EuOCP3 as a functional ingredient in food products for anti-OP treatment.
Collapse
Affiliation(s)
- Jiyu Song
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yongfeng Zhang
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yutong Li
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
35
|
Viegas C, Carreira J, Maia TM, Macedo AL, Matos AP, Neves J, Simes D. Gla Rich Protein (GRP) Mediates Vascular Smooth Muscle Cell (VSMC) Osteogenic Differentiation, Extracellular Vesicle (EV) Calcification Propensity, and Immunomodulatory Properties. Int J Mol Sci 2024; 25:12406. [PMID: 39596469 PMCID: PMC11594964 DOI: 10.3390/ijms252212406] [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: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Vascular calcification (VC) is a complex process involving vascular smooth muscle cell (VSMC) osteogenic differentiation, inflammation, and extracellular vesicle (EV) calcification and communication networks. Gla rich protein (GRP) is a calcification inhibitor involved in most of these processes. However, the molecular mechanism of GRP in VC and the specific characteristics, cargo, and functionality of calcifying EVs require further elucidation. Here, we use a combination of human ex vivo aortic fragments and primary vascular smooth muscle cell (VSMC) models to obtain new information on GRP function in VC and EVs released by VSMCs. We demonstrate that GRP inhibits VSMC osteogenic differentiation through downregulation of bone-related proteins and upregulation of mineralization inhibitors, with decreased mineral crystallinity in EVs deposited into the tissue extracellular matrix (ECM). EVs isolated by ultracentrifugation at 30K and 100K from the cell media (CM) and deposited in the ECM from control (CTR) and mineralizing (MM) VSMCs were biochemically, physically, and proteomically characterized. Four different EV populations were identified with shared markers commonly present in all EVs but with unique protein cargo and specific molecular profiles. Comparative proteomics identified several regulated proteins specifically loaded into MM EV populations associated with multiple processes involved in VC. Functional analysis demonstrated that 30K and 100K ECM-MM EVs with higher calcium and lower GRP levels induced macrophage inflammation. Our findings reinforce the functional relevance of GRP in multiple VC processes and suggest that ECM EVs released under calcification stress function as a new signaling axis on the calcification-inflammation cycle.
Collapse
Affiliation(s)
- Carla Viegas
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| | - Joana Carreira
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
| | - Teresa M. Maia
- VIB Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
- VIB Proteomics Core, 9052 Ghent, Belgium
| | - Anjos L. Macedo
- UCIBIO, Department of Chemistry, and Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - António P. Matos
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, 2829-511 Caparica, Portugal;
| | - José Neves
- Service of Cardiothoracic Surgery, Santa Cruz Hospital, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal;
| | - Dina Simes
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
36
|
Kabir A, B M, A N, Selvaraj V, Sudhakar S. Protein Nano Coop Complexes Promote Fracture Healing and Bone Regeneration in a Zebrafish Osteoporosis Model. Biomacromolecules 2024; 25:7237-7248. [PMID: 39449233 DOI: 10.1021/acs.biomac.4c00931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Nanotherapeutic techniques are becoming increasingly important in the treatment of bone disorders owing to their targeted drug delivery. This study formulates zein nano coop composites containing chimeric antioxidants (ascorbic acid, luteolin, resveratrol, and coenzyme Q) (AZN) and evaluates its application in bone regeneration using osteoblasts and a Zebrafish osteoporosis model. In vitro experiments with human osteoblast-like MG63 cells showed enhancement of bone mineralization and regeneration. It further exhibited high biocompatibility in Zebrafish larvae, with increased calcium/phosphorus deposition and upregulation of osteogenic genes. The study has unequivocally demonstrated the potential of AZN in bone regeneration and fracture healing in both normal and osteoporosis models, underscoring the significance of this research. Further investigations using higher animal models are warranted to expand on these findings. The impact of this research seems far-reaching, with the possible development of new, effective, and safe treatment options for osteoporosis, addressing the limitations of the currently available treatments.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Mukilarasi B
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Natarajan A
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| |
Collapse
|
37
|
Xinyi Y, Vladimirovich RI, Beeraka NM, Satyavathi A, Kamble D, Nikolenko VN, Lakshmi AN, Basappa B, Reddy Y P, Fan R, Liu J. Emerging insights into epigenetics and hematopoietic stem cell trafficking in age-related hematological malignancies. Stem Cell Res Ther 2024; 15:401. [PMID: 39506818 PMCID: PMC11539620 DOI: 10.1186/s13287-024-04008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Hematopoiesis within the bone marrow (BM) is a complex and tightly regulated process predominantly influenced by immune factors. Aging, diabetes, and obesity are significant contributors to BM niche damage, which can alter hematopoiesis and lead to the development of clonal hematopoiesis of intermediate potential (CHIP). Genetic/epigenetic alterations during aging could influence BM niche reorganization for hematopoiesis or clonal hematopoiesis. CHIP is driven by mutations in genes such as Tet2, Dnmt3a, Asxl1, and Jak2, which are associated with age-related hematological malignancies. OBJECTIVE This literature review aims to provide an updated exploration of the functional aspects of BM niche cells within the hematopoietic microenvironment in the context of age-related hematological malignancies. The review specifically focuses on how immunological stressors modulate different signaling pathways that impact hematopoiesis. METHODS An extensive review of recent studies was conducted, examining the roles of various BM niche cells in hematopoietic stem cell (HSC) trafficking and the development of age-related hematological malignancies. Emphasis was placed on understanding the influence of immunological stressors on these processes. RESULTS Recent findings reveal a significant microheterogeneity and temporal stochasticity of niche cells across the BM during hematopoiesis. These studies demonstrate that niche cells, including mesenchymal stem cells, osteoblasts, and endothelial cells, exhibit dynamic interactions with HSCs, significantly influenced by the BM microenvironment as the age increases. Immunosurveillance plays a crucial role in maintaining hematopoietic homeostasis, with alterations in immune signaling pathways contributing to the onset of hematological malignancies. Novel insights into the interaction between niche cells and HSCs under stress/aging conditions highlight the importance of niche plasticity and adaptability. CONCLUSION The involvement of age-induced genetic/epigenetic alterations in BM niche cells and immunological stressors in hematopoiesis is crucial for understanding the development of age-related hematological malignancies. This comprehensive review provides new insights into the complex interplay between niche cells and HSCs, emphasizing the potential for novel therapeutic approaches that target niche cell functionality and resilience to improve hematopoietic outcomes in the context of aging and metabolic disorders. NOVELTY STATEMENT This review introduces novel concepts regarding the plasticity and adaptability of BM niche cells in response to immunological stressors and epigenetics. It proposes that targeted therapeutic strategies aimed at enhancing niche cell resilience could mitigate the adverse effects of aging, diabetes, and obesity on hematopoiesis and clonal hematopoiesis. Additionally, the review suggests that understanding the precise temporal and spatial dynamics of niche-HSC interactions and epigenetics influence may lead to innovative treatments for age-related hematological malignancies.
Collapse
Affiliation(s)
- Yang Xinyi
- Department of Oncology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Reshetov Igor Vladimirovich
- Department of Oncology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia.
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India.
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA.
- Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Allaka Satyavathi
- Department of Chemistry, Faculty of science, Dr B R Ambedkar Open University, Wanaparthy, Telangana, 509103, India
| | - Dinisha Kamble
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Allaka Naga Lakshmi
- Department of Computer Science, St Philomena's College (Autonomous), Bangalore - Mysore Rd, Bannimantap, Mysuru, Karnataka, 570015, India
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, Karnataka, 570006, India
| | - Padmanabha Reddy Y
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, China.
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, China
| |
Collapse
|
38
|
Azarkina K, Gromova E, Malashicheva A. "A Friend Among Strangers" or the Ambiguous Roles of Runx2. Biomolecules 2024; 14:1392. [PMID: 39595568 PMCID: PMC11591759 DOI: 10.3390/biom14111392] [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: 09/01/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The transcription factor Runx2 plays a crucial role in regulating osteogenic differentiation and skeletal development. This factor not only controls the expression of genes involved in bone formation, but also interacts with signaling pathways such as the Notch pathway, which are essential for body development. However, studies have produced conflicting results regarding the relationship between Runx2 and the Notch pathway. Some studies suggest a synergistic interaction between these molecules, while others suggest an inhibitory one, for example, the interplay between Notch signaling, Runx2, and vitamin D3 in osteogenic differentiation and bone remodeling. The findings suggest a complex relationship between Notch signaling and osteogenic differentiation, with ongoing research needed to clarify the mechanisms involved and resolve existing contradictions regarding role of Notch in this process. Additionally, there is increasing evidence of contradictory roles for Runx2 in various tissues and organs, both under normal conditions and in pathological states. This diversity of roles makes Runx2 a potential therapeutic target, offering new directions for research. In this review, we have discussed the mechanisms of osteogenic differentiation and the important role of Runx2 in this process. We have also examined its relationship with different signaling pathways. However, there are still many uncertainties and inconsistencies in our current understanding of these interactions. Additionally, given that Runx2 is also involved in numerous other events in various tissues, we have tried to comprehensively examine its functions outside the skeletal system.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, 194064 Saint-Petersburg, Russia
| |
Collapse
|
39
|
Zhang C, Chang Y, Shu L, Chen Z. Pathogenesis of thoracic ossification of the ligamentum flavum. Front Pharmacol 2024; 15:1496297. [PMID: 39545059 PMCID: PMC11560781 DOI: 10.3389/fphar.2024.1496297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Thoracic ossification of the ligamentum flavum (TOLF) is characterized by ectopic ossification of the ligamentum flavum in the thoracic spine and is considered the main cause of thoracic spinal stenosis and spinal cord disease. Osteoblast specific transcription factor Osterix (Osx) is required for bone formation, and there is no bone formation or ossification without Osx. Surgical intervention is recognized as the only effective method for TOLF treatment with set of complications. However, underlying mechanisms of TOLF are not well understood. This paper summarizes the pathogenesis of TOLF. Some relevant factors have been discussed, such as mechanical stress, genetic susceptibility genes, endocrine and trace element metabolism abnormalities, which may associate with TOLF. More recent studies using proteomics technology and RNA sequencing approach have discovered that some new factors participate in TOLF by upregulation of Osx gene expression including inflammatory factors. TOLF is a unique disease involving multiple factors. On the other hand, studies on TOLF pathogenic mechanism may provide new ideas for finding possible upstream regulatory factors of Osx and further developing novel drugs to stimulate new bone formation to treat osteoporosis.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, Peking University International Hospital, Beijing, China
- Central Laboratory, Peking University International Hospital, Beijing, China
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yanan Chang
- Central Laboratory, Peking University International Hospital, Beijing, China
| | - Li Shu
- Central Laboratory, Peking University International Hospital, Beijing, China
| | - Zhongqiang Chen
- Department of Orthopedics, Peking University International Hospital, Beijing, China
| |
Collapse
|
40
|
German IJS, Barbalho SM, Andreo JC, Zutin TLM, Laurindo LF, Rodrigues VD, Araújo AC, Guiguer EL, Direito R, Pomini KT, Shinohara AL. Exploring the Impact of Catechins on Bone Metabolism: A Comprehensive Review of Current Research and Future Directions. Metabolites 2024; 14:560. [PMID: 39452941 PMCID: PMC11509841 DOI: 10.3390/metabo14100560] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Degenerative musculoskeletal diseases represent a global health problem due to the progressive deterioration of affected individuals. As a bioactive compound, catechins have shown osteoprotective properties by stimulating osteoblastic cells and inhibiting bone resorption. Thus, this review aimed to address the mechanism of action of catechins on bone tissue. Methods: The search was applied to PubMed without limitations in date, language, or article type. Fifteen articles matched the topic and objective of this review. Results: EGCG (epigallocatechin gallate) and epicatechin demonstrated action on the osteogenic markers RANKL, TRAP, and NF-κβ and expression of BMPs and ALP, thus improving the bone microarchitecture. Studies on animals showed the action of EGCG in increasing calcium and osteoprotegerin levels, in addition to regulating the transcription factor NF-ATc1 associated with osteoclastogenesis. However, it did not show any effect on osteocalcin and RANK. Regarding human studies, EGCG reduced the risk of fracture in a dose-dependent manner. In periodontal tissue, EGCG reduced IL-6, TNF, and RANKL in vitro and in vivo. Human studies showed a reduction in periodontal pockets, gingival index, and clinical attachment level. The action of EGCG on membranes and hydrogels showed biocompatible and osteoinductive properties on the microenvironment of bone tissue by stimulating the expression of osteogenic growth factors and increasing osteocalcin and alkaline phosphate levels, thus promoting new bone formation. Conclusions: EGCG stimulates cytokines related to osteogenes, increasing bone mineral density, reducing osteoclastogenesis factors, and showing great potential as a therapeutic strategy for reducing the risk of bone fractures.
Collapse
Affiliation(s)
- Iris Jasmin Santos German
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| | - Sandra Maria Barbalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Research Coordination, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Jesus Carlos Andreo
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| | - Tereza Lais Menegucci Zutin
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, Brazil; (L.F.L.)
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, Brazil; (L.F.L.)
| | - Adriano Cressoni Araújo
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Elen Landgraf Guiguer
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed. ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Karina Torres Pomini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - André Luis Shinohara
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| |
Collapse
|
41
|
Bertels JC, He G, Long F. Metabolic reprogramming in skeletal cell differentiation. Bone Res 2024; 12:57. [PMID: 39394187 PMCID: PMC11470040 DOI: 10.1038/s41413-024-00374-0] [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: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
The human skeleton is a multifunctional organ made up of multiple cell types working in concert to maintain bone and mineral homeostasis and to perform critical mechanical and endocrine functions. From the beginning steps of chondrogenesis that prefigures most of the skeleton, to the rapid bone accrual during skeletal growth, followed by bone remodeling of the mature skeleton, cell differentiation is integral to skeletal health. While growth factors and nuclear proteins that influence skeletal cell differentiation have been extensively studied, the role of cellular metabolism is just beginning to be uncovered. Besides energy production, metabolic pathways have been shown to exert epigenetic regulation via key metabolites to influence cell fate in both cancerous and normal tissues. In this review, we will assess the role of growth factors and transcription factors in reprogramming cellular metabolism to meet the energetic and biosynthetic needs of chondrocytes, osteoblasts, or osteoclasts. We will also summarize the emerging evidence linking metabolic changes to epigenetic modifications during skeletal cell differentiation.
Collapse
Affiliation(s)
- Joshua C Bertels
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guangxu He
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedics, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Fanxin Long
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Doyle SE, Cazzola CN, Coleman CM. Design considerations when creating a high throughput screen-compatible in vitro model of osteogenesis. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100184. [PMID: 39313131 DOI: 10.1016/j.slasd.2024.100184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Inducing osteogenic differentiation in vitro is useful for the identification and development of bone regeneration therapies as well as modelling bone disorders. To couple in vitro models with high throughput screening techniques retains the assay's relevance in research while increasing its therapeutic impact. Miniaturizing, automating and/or digitalizing in vitro assays will reduce the required quantity of cells, biologic stimulants, culture/output assay reagents, time and cost. This review highlights the design and workflow considerations for creating a high throughput screen-compatible model of osteogenesis, comparing and contrasting osteogenic cell type, assay fabrication and culture methodology, osteogenic induction approach and repurposing existing output techniques.
Collapse
Affiliation(s)
- Stephanie E Doyle
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland.
| | - Courtney N Cazzola
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| | - Cynthia M Coleman
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| |
Collapse
|
43
|
Gomez GA, Udayakumar A, Pourteymoor S, Dennis G, Xing W, Mohan S. Evaluation of Potential Roles of Zinc Finger Homeobox 3 (Zfhx3) Expressed in Chondrocytes and Osteoblasts on Skeletal Growth in Mice. Calcif Tissue Int 2024; 115:445-454. [PMID: 39085428 PMCID: PMC11648307 DOI: 10.1007/s00223-024-01265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024]
Abstract
Bone formation is tightly modulated by genetically encoded molecular proteins that interact to regulate cellular differentiation and secretion of bony matrix. Many transcription factors are known to coordinate these events by controlling gene transcription within networks. However, not all factors involved are known. Here, we identified a novel function for Zinc Finger Homeobox 3 (Zfhx3), a gene encoding a transcription factor, as a regulator of bone metabolism. We knocked out Zfhx3 conditionally in mice in either chondrocytes or osteoblasts and characterized their bones by micro-CT in 12-week-old mice. We observed a negative effect in linear bone growth in both knockout mice but reduced bone mass only in mice with Zfhx3 deleted in osteoblasts. Loss of Zfhx3 expression in osteoblasts affected trabecular bone mass in femurs and vertebrae in both sexes but influenced cortical bone volume fraction only in females. Moreover, transcriptional analysis of femoral bones in osteoblast Zfhx3 conditional knockout mice revealed a reduced expression of osteoblast genes, and histological evaluation of trabecular bones suggests that Zfhx3 causes changes in bone formation and not resorption. The loss of Zfhx3 causes reductions in trabecular bone area and osteoid volume, but no changes in the expression of osteoclast differentiation markers or number of TRAP stained osteoclasts. These studies introduce Zfhx3 as a relevant factor toward understanding gene regulatory networks that control bone formation and development of peak bone mass.
Collapse
Affiliation(s)
- Gustavo A Gomez
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | - Anakha Udayakumar
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | - Garrett Dennis
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | - Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA.
- Departments of Medicine, Biochemistry and Orthopedic Surgery, Loma Linda University, Loma Linda, CA, 92354, USA.
| |
Collapse
|
44
|
Wang L, Zheng G, Yuan Y, Wang Z, Wang Q, Sun M, Wu J, Liu C, Liu Y, Zhang B, Zhang H, Yang N, Lian L. circRUNX2.2, highly expressed in Marek's disease tumor tissues, functions in cis to regulate parental gene RUNX2 expression. Poult Sci 2024; 103:104045. [PMID: 39094493 PMCID: PMC11345620 DOI: 10.1016/j.psj.2024.104045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Marek's disease (MD), an immunosuppression disease induced by Marek's disease virus (MDV), is one of the significant diseases affecting the health and productive performance of poultry. The roles of circular RNAs (circRNAs) in MD development were poorly understood. In this study, we found a circRNA derived from exon 6 of RUNX family transcription factor 2 (RUNX2) gene, named circRUNX2.2, was highly expressed in chicken tumorous spleens (TS) induced by MDV. Through fluorescence in situ hybridization and nuclear-cytoplasmic separation assay, we determined circRUNX2.2 was mainly located in the nucleus. Knockout experiments confirmed that the flanking complementary sequences (RCMs) mediated its circularization. Gain of function assay and dual luciferase reporter gene assay revealed that circRUNX2.2 could promote the expression of RUNX2 via binding with its promoter region. RNA antisense purification assay and mass spectrometry assay showed circRUNX2.2 could recruit proteins such as CHD9 protein. Knocking down CHD9 expression decreased the expression of RUNX2 gene, which confirmed the positive regulation that circRUNX2.2 on RUNX2 expression was probably facilitated via recruiting CHD9 protein. Functional experiments showed that circRUNX2.2 promoted the proliferation of the MD lymphoma-derived chicken cell line, MDCC-MSB1, which confirmed the potential oncogenic role of circRNX2.2 in tumor development. In conclusion, we found that the RUNX2-derived circRUNX2.2 can positively regulate the transcription of the parental gene RUNX2 in a cis-acting manner. The high expression of circRUNX2.2 in MD tumor tissues indicated that it might mediate MD lymphoma progression.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Gang Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yiming Yuan
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ziyi Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qinyuan Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Meng Sun
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junfeng Wu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Changjun Liu
- Division of Avian Infectious Diseases, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yongzhen Liu
- Division of Avian Infectious Diseases, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Bo Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hao Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ling Lian
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
45
|
Platko K, Gyulay G, Lebeau PF, MacDonald ME, Lynn EG, Byun JH, Igdoura SA, Holden RM, Roubtsova A, Seidah NG, Krepinsky JC, Austin RC. GDF10 is a negative regulator of vascular calcification. J Biol Chem 2024; 300:107805. [PMID: 39307303 PMCID: PMC11541827 DOI: 10.1016/j.jbc.2024.107805] [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: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 10/27/2024] Open
Abstract
Cardiovascular mortality is particularly high and increasing in patients with chronic kidney disease, with vascular calcification (VC) as a major pathophysiologic feature. VC is a highly regulated biological process similar to bone formation involving osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). We have previously demonstrated that loss of T-cell death-associated gene 51 (TDAG51) expression leads to an attenuation of medial VC. We now show a significant induction of circulating levels of growth differentiation factor 10 (GDF10) in TDAG51-/- mice, which was of interest due to its established role as an inhibitor of osteoblast differentiation. The objective of this study was to examine the role of GDF10 in the osteogenic transdifferentiation of VSMCs. Using primary mouse and human VSMCs, as well as ex vivo aortic ring cultures, we demonstrated that treatment with recombinant human (rh) GDF10 mitigated phosphate-mediated hydroxyapatite (HA) mineral deposition. Furthermore, ex vivo aortic rings from GDF10-/- mice exhibited increased HA deposition compared to C57BL/6J controls. To explain our observations, we identified that rhGDF10 treatment reduced protein expression of runt-related transcription factor 2, a key driver of osteogenic transdifferentiation of VSMCs and VC. In support of these findings, in vivo treatment with rhGDF10 attenuated VD3-induced VC. Furthermore, we demonstrated an increase in circulating GDF10 in patients with chronic kidney disease with clinically defined severe VC, as assessed by coronary artery calcium score. Thus, our studies identify GDF10 as a novel inhibitor of mineral deposition and as such, may represent a potential novel biomarker and therapeutic target for the detection and management of VC.
Collapse
Affiliation(s)
- Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Gabriel Gyulay
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Melissa E MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Suleiman A Igdoura
- Department of Biology, McMaster University Medical Centre, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Anna Roubtsova
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Nabil G Seidah
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Joan C Krepinsky
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| |
Collapse
|
46
|
Komori T. Regulation of Skeletal Development and Maintenance by Runx2 and Sp7. Int J Mol Sci 2024; 25:10102. [PMID: 39337587 PMCID: PMC11432631 DOI: 10.3390/ijms251810102] [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: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Runx2 (runt related transcription factor 2) and Sp7 (Sp7 transcription factor 7) are crucial transcription factors for bone development. The cotranscription factor Cbfb (core binding factor beta), which enhances the DNA-binding capacity of Runx2 and stabilizes the Runx2 protein, is necessary for bone development. Runx2 is essential for chondrocyte maturation, and Sp7 is partly involved. Runx2 induces the commitment of multipotent mesenchymal cells to osteoblast lineage cells and enhances the proliferation of osteoprogenitors. Reciprocal regulation between Runx2 and the Hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathways and Dlx5 (distal-less homeobox 5) plays an important role in these processes. The induction of Fgfr2 (Fgf receptor 2) and Fgfr3 expression by Runx2 is important for the proliferation of osteoblast lineage cells. Runx2 induces Sp7 expression, and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Sp7 induces the differentiation of preosteoblasts into osteoblasts without enhancing their proliferation. In osteoblasts, Runx2 is required for bone formation by inducing the expression of major bone matrix protein genes, including Col1a1 (collagen type I alpha 1), Col1a2, Spp1 (secreted phosphoprotein 1), Ibsp (integrin binding sialoprotein), and Bglap (bone gamma carboxyglutamate protein)/Bglap2. Bglap/Bglap2 (osteocalcin) regulates the alignment of apatite crystals parallel to collagen fibrils but does not function as a hormone that regulates glucose metabolism, testosterone synthesis, and muscle mass. Sp7 is also involved in Co1a1 expression and regulates osteoblast/osteocyte process formation, which is necessary for the survival of osteocytes and the prevention of cortical porosity. SP7 mutations cause osteogenesis imperfecta in rare cases. Runx2 is an important pathogenic factor, while Runx1, Runx3, and Cbfb are protective factors in osteoarthritis development.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
47
|
Freiberger RN, López CAM, Palma MB, Cevallos C, Sviercz FA, Jarmoluk P, García MN, Quarleri J, Delpino MV. HIV Modulates Osteoblast Differentiation via Upregulation of RANKL and Vitronectin. Pathogens 2024; 13:800. [PMID: 39338991 PMCID: PMC11435243 DOI: 10.3390/pathogens13090800] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Bone loss is a prevalent characteristic among people with HIV (PWH). We focused on mesenchymal stem cells (MSCs) and osteoblasts, examining their susceptibility to different HIV strains (R5- and X4-tropic) and the subsequent effects on bone tissue homeostasis. Our findings suggest that MSCs and osteoblasts are susceptible to R5- and X4-tropic HIV but do not support productive HIV replication. HIV exposure during the osteoblast differentiation process revealed that the virus could not alter mineral and organic matrix deposition. However, the reduction in runt-related transcription factor 2 (RUNX2) transcription, the increase in the transcription of nuclear receptor activator ligand kappa B (RANKL), and the augmentation of vitronectin deposition strongly suggested that X4- and R5-HIV could affect bone homeostasis. This study highlights the HIV ability to alter MSCs' differentiation into osteoblasts, critical for maintaining bone and adipose tissue homeostasis and function.
Collapse
Affiliation(s)
- Rosa Nicole Freiberger
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - Cynthia Alicia Marcela López
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - María Belén Palma
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fleni, Consejo de Investigaciones Científicas y Técnicas (CONICET), Escobar 1625, Argentina
| | - Cintia Cevallos
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - Franco Agustin Sviercz
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - Patricio Jarmoluk
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - Marcela Nilda García
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Jorge Quarleri
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| | - M. Victoria Delpino
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina; (R.N.F.); (C.A.M.L.); (C.C.); (P.J.); (J.Q.)
| |
Collapse
|
48
|
Onoki T, Kanczler J, Rawlings A, Smith M, Kim YH, Hashimoto K, Aizawa T, Oreffo ROC. Modulation of osteoblastogenesis by NRF2: NRF2 activation suppresses osteogenic differentiation and enhances mineralization in human bone marrow-derived mesenchymal stromal cells. FASEB J 2024; 38:e23892. [PMID: 39230563 DOI: 10.1096/fj.202400602r] [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: 03/18/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
Mesenchymal stromal stem cells (MSCs) or skeletal stem cells (SSCs) play a major role in tissue repair due to their robust ability to differentiate into osteoblasts, chondrocytes, and adipocytes. Complex cell signaling cascades tightly regulate this differentiation. In osteogenic differentiation, Runt-related transcription factor 2 (RUNX2) and ALP activity are essential. Furthermore, during the latter stages of osteogenic differentiation, mineral formation mediated by the osteoblast occurs with the secretion of a collagenous extracellular matrix and calcium deposition. Activation of nuclear factor erythroid 2-related factor 2 (NRF2), an important transcription factor against oxidative stress, inhibits osteogenic differentiation and mineralization via modulation of RUNX2 function; however, the exact role of NRF2 in osteoblastogenesis remains unclear. Here, we demonstrate that NRF2 activation in human bone marrow-derived stromal cells (HBMSCs) suppressed osteogenic differentiation. NRF2 activation increased the expression of STRO-1 and KITLG (stem cell markers), indicating NRF2 protects HBMSCs stemness against osteogenic differentiation. In contrast, NRF2 activation enhanced mineralization, which is typically linked to osteogenic differentiation. We determined that these divergent results were due in part to the modulation of cellular calcium flux genes by NRF2 activation. The current findings demonstrate a dual role for NRF2 as a HBMSC maintenance factor as well as a central factor in mineralization, with implications therein for elucidation of bone formation and cellular Ca2+ kinetics, dystrophic calcification and, potentially, application in the modulation of bone formation.
Collapse
Affiliation(s)
- Takahiro Onoki
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Andrew Rawlings
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Melanie Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Ko Hashimoto
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Toshimi Aizawa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
49
|
Upadhyay V, Sharma S, Sethi A, Singh AK, Chowdhury S, Srivastava S, Mishra S, Singh S, Chattopadhyay N, Trivedi AK. Hakai, a novel Runx2 interacting protein, augments osteoblast differentiation by rescuing Runx2 from Smurf2-mediated proteasome degradation. J Cell Physiol 2024; 239:e31388. [PMID: 39034451 DOI: 10.1002/jcp.31388] [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: 04/18/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Runt-related transcription factor 2 (Runx2) is a key regulator of osteoblast differentiation and bone formation. In Runx2-deficient embryos, skeletal development ceases at the cartilage anlage stage. These embryos die of respiratory failure upon birth and display a complete absence of bone and cartilage mineralization. Here, we identified Hakai, a type of E3 ubiquitin ligase as a potential Runx2 interacting partner through affinity pulldown-based proteomic approach. Subsequently, we observed that similar to Runx2, Hakai was downregulated in osteopenic ovariectomized rats, suggesting its involvement in bone formation. Consistent with this observation, Hakai overexpression significantly enhanced osteoblast differentiation in mesenchyme-like C3H10T1/2 as well as primary rat calvaria osteoblast (RCO) cells in vitro. Conversely, overexpression of a catalytically inactive Hakai mutant (C109A) exhibited minimal to no effect, whereas Hakai depletion markedly reduced endogenous Runx2 levels and impaired osteogenic differentiation in both C3H10T1/2 and RCOs. Mechanistically, Hakai physically interacts with Runx2 and enhances its protein turnover by rescuing it from Smad ubiquitination regulatory factor 2 (Smurf2)-mediated proteasome degradation. Wild-type Hakai but not Hakai-C109A inhibited Smurf2 protein levels through proteasome-mediated degradation. These findings underscore Hakai's functional role in bone formation, primarily through its positive modulation of Runx2 protein turnover by protecting it from Smurf2-mediated ubiquitin-proteasomal degradation. Collectively, our results demonstrate Hakai as a promising novel therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Vishal Upadhyay
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Shivani Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
- Division of Endocrinology and Center for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Arppita Sethi
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anil Kumar Singh
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Sangita Chowdhury
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Swati Srivastava
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Shivkant Mishra
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Shyam Singh
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
- Division of Endocrinology and Center for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Arun Kumar Trivedi
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
50
|
Castillo H, Hanna P, Sachs LM, Buisine N, Godoy F, Gilbert C, Aguilera F, Muñoz D, Boisvert C, Debiais-Thibaud M, Wan J, Spicuglia S, Marcellini S. Xenopus tropicalis osteoblast-specific open chromatin regions reveal promoters and enhancers involved in human skeletal phenotypes and shed light on early vertebrate evolution. Cells Dev 2024; 179:203924. [PMID: 38692409 DOI: 10.1016/j.cdev.2024.203924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
While understanding the genetic underpinnings of osteogenesis has far-reaching implications for skeletal diseases and evolution, a comprehensive characterization of the osteoblastic regulatory landscape in non-mammalian vertebrates is still lacking. Here, we compared the ATAC-Seq profile of Xenopus tropicalis (Xt) osteoblasts to a variety of non mineralizing control tissues, and identified osteoblast-specific nucleosome free regions (NFRs) at 527 promoters and 6747 distal regions. Sequence analyses, Gene Ontology, RNA-Seq and ChIP-Seq against four key histone marks confirmed that the distal regions correspond to bona fide osteogenic transcriptional enhancers exhibiting a shared regulatory logic with mammals. We report 425 regulatory regions conserved with human and globally associated to skeletogenic genes. Of these, 35 regions have been shown to impact human skeletal phenotypes by GWAS, including one trps1 enhancer and the runx2 promoter, two genes which are respectively involved in trichorhinophalangeal syndrome type I and cleidocranial dysplasia. Intriguingly, 60 osteoblastic NFRs also align to the genome of the elephant shark, a species lacking osteoblasts and bone tissue. To tackle this paradox, we chose to focus on dlx5 because its conserved promoter, known to integrate regulatory inputs during mammalian osteogenesis, harbours an osteoblast-specific NFR in both frog and human. Hence, we show that dlx5 is expressed in Xt and elephant shark odontoblasts, supporting a common cellular and genetic origin of bone and dentine. Taken together, our work (i) unravels the Xt osteogenic regulatory landscape, (ii) illustrates how cross-species comparisons harvest data relevant to human biology and (iii) reveals that a set of genes including bnc2, dlx5, ebf3, mir199a, nfia, runx2 and zfhx4 drove the development of a primitive form of mineralized skeletal tissue deep in the vertebrate lineage.
Collapse
Affiliation(s)
- Héctor Castillo
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile.
| | - Patricia Hanna
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Laurent M Sachs
- UMR7221, Physiologie Moléculaire et Adaptation, CNRS, MNHN, Paris Cedex 05, France
| | - Nicolas Buisine
- UMR7221, Physiologie Moléculaire et Adaptation, CNRS, MNHN, Paris Cedex 05, France
| | - Francisco Godoy
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Clément Gilbert
- Université Paris-Saclay, CNRS, IRD, UMR Évolution, Génomes, Comportement et Écologie, 12 route 128, 91190 Gif-sur-Yvette, France
| | - Felipe Aguilera
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - David Muñoz
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Catherine Boisvert
- School of Molecular and Life Sciences, Curtin University, Perth, WA, Australia
| | - Mélanie Debiais-Thibaud
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Univ Montpellier, CNRS, IRD, Montpellier, France
| | - Jing Wan
- Aix-Marseille University, INSERM, TAGC, UMR 1090, Marseille, France; Equipe Labelisée LIGUE contre le Cancer, Marseille, France
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR 1090, Marseille, France; Equipe Labelisée LIGUE contre le Cancer, Marseille, France
| | - Sylvain Marcellini
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile.
| |
Collapse
|