1
|
Rillaerts K, Verlinden L, Doms S, Carmeliet G, Verstuyf A. A comprehensive perspective on the role of vitamin D signaling in maintaining bone homeostasis: Lessons from animal models. J Steroid Biochem Mol Biol 2025; 250:106732. [PMID: 40122304 DOI: 10.1016/j.jsbmb.2025.106732] [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: 12/16/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
1,25(OH)2D3 is well known for its role in maintaining normal serum calcium levels. Through its receptor, 1,25(OH)2D3 enhances intestinal calcium absorption and renal calcium reabsorption, thereby ensuring serum calcium levels are within physiological range, which is in turn important for normal bone development and mineralization. The vitamin D receptor (VDR) achieves this via transcriptional induction of genes important in calcium transport. When intestinal and renal calcium (re)absorption is impaired, VDR-mediated signaling will stimulate bone resorption and inhibit mineralization in order to maintain normal serum calcium levels, as evidenced in mice with a systemic or intestine-specific deletion of the VDR. However, VDR signaling in bone is also reported to have anabolic effects. In this review we will discuss the effects of 1,25(OH)2D3-mediated VDR signaling on bone homeostasis and provide an overview of the in vitro experiments and various transgenic mice models that have been generated to unravel the role of VDR signaling in different bone cell types such as chondrocytes, (pre)osteoblasts, osteocytes, and (pre)osteoclasts.
Collapse
Affiliation(s)
- Kayleigh Rillaerts
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Lieve Verlinden
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Stefanie Doms
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| | - Annemieke Verstuyf
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, bus 902, Leuven 3000, Belgium
| |
Collapse
|
2
|
Weinhaus B, Homan S, Kincaid M, Tadwalkar A, Gu X, Kumar S, Slaughter A, Zhang J, Wu Q, Kofron JM, Troutman TD, DeFalco T, Lucas D. Differential regulation of fetal bone marrow and liver hematopoiesis by yolk-sac-derived myeloid cells. Nat Commun 2025; 16:4427. [PMID: 40368906 PMCID: PMC12078524 DOI: 10.1038/s41467-025-59058-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Fetal hematopoiesis takes place in the liver before colonizing the bone marrow where it will persist for life. This colonization is thought to be mediated by specification of a microenvironment that selectively recruits hematopoietic cells to the nascent bone marrow. The identity and mechanisms regulating the specification of this colonization niche are unclear. Here we identify a VCAM1+ sinusoidal colonization niche in the diaphysis that regulates neutrophil and hematopoietic stem cell colonization of the bone marrow. Using confocal microscopy, we find that colonizing hematopoietic stem and progenitor cells (HSPC) and myeloid cells selectively localize to a subset of VCAM1+ sinusoids in the center of the diaphysis. Vcam1 deletion in endothelial cells impairs hematopoietic colonization while depletion of yolk-sac-derived osteoclasts disrupts VCAM1+ expression, and impairs neutrophil and HSPC colonization to the bone marrow. Depletion of yolk-sac-derived myeloid cells increases fetal liver hematopoietic stem cell numbers, function and erythropoiesis independent of osteoclast activity. Thus, the yolk sac produces myeloid cells that have opposite roles in fetal hematopoiesis: while yolk-sac derived myeloid cells in the bone marrow promote hematopoietic colonization by specifying a VCAM1+ colonization niche, a different subset of yolk-sac-derived myeloid cells inhibits HSC in the fetal liver.
Collapse
Affiliation(s)
- Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Shelli Homan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Morgan Kincaid
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Aryan Tadwalkar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Sumit Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Anastasiya Slaughter
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Jizhou Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Qingqing Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ty D Troutman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
3
|
Garmendia Urdalleta A, Witte-Bouma J, Kops N, Lolli A, Farrell E. Osteoclast Incorporation in an In Vitro 3D Model of Endochondral Ossification. Tissue Eng Part A 2025. [PMID: 40279297 DOI: 10.1089/ten.tea.2024.0281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025] Open
Abstract
In vitro models aim to recapitulate human physiological processes, improving upon and replacing the need for animal-based models. Modeling bone formation via endochondral ossification in vitro is a very complex process due to the large number of cell types involved. Most current models are limited to mimicking the initial stages of the process (i.e., cartilage template formation and mineralization of the matrix), using a single cell type. Chondroclasts/osteoclasts are key players in cartilage resorption during endochondral ossification, but their introduction into in vitro models has thus far proven challenging. In this study, we aimed toward a new level of model complexity by introducing human monocyte-derived osteoclasts into 3D in vitro-cultured cartilage templates undergoing mineralization. Chondrogenic and mineralized chondrogenic pellets were formed from human pediatric bone marrow stromal cells and cultured in the presence of transforming growth factor-β3 (TGF-β) and TGF-β/β-glycerophosphate, respectively. These pellets have the capacity to form bone if implanted in vivo. To identify suitable in vitro co-culture conditions and investigate cell interactions, pellets were co-cultured with CD14+ monocytes in an indirect (transwell) or direct setting for up to 14 days, and osteoclastogenesis was assessed by means of histological stainings, osteoclast counting, and gene expression analysis. Upon direct co-culture, we achieved effective osteoclast formation in situ in regions of both mineralized and unmineralized cartilages. Notably, in vitro-generated osteoclasts showed the ability to form tunnels in the chondrogenic matrix and infiltrate the mineralized matrix. Addition of osteoclasts in human in vitro models of endochondral ossification increases the physiological relevance of these models. This will allow for the development of robust 3D human in vitro systems for the study of bone formation, disease modeling, and drug discovery, further reducing the need for animal models in the future.
Collapse
Affiliation(s)
- Amaia Garmendia Urdalleta
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Shi D, Li Y, Tian M, Xue M, Wang J, An H. Nanomaterials-Based Drug Delivery Systems for Therapeutic Applications in Osteoporosis. Adv Biol (Weinh) 2025:e2400721. [PMID: 40195930 DOI: 10.1002/adbi.202400721] [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: 11/19/2024] [Revised: 03/04/2025] [Indexed: 04/09/2025]
Abstract
The etiology of osteoporosis is rooted in the disruption of the intricate equilibrium between bone formation and bone resorption processes. Nevertheless, the conventional anti-osteoporotic medications and hormonal therapeutic regimens currently employed in clinical practice are associated with a multitude of adverse effects, thereby constraining their overall therapeutic efficacy and potential. Recently, nanomaterials have emerged as a promising alternative due to their minimal side effects, efficient drug delivery, and ability to enhance bone formation, aiding in restoring bone balance. This review delves into the fundamental principles of bone remodeling and the bone microenvironment, as well as current clinical treatment approaches for osteoporosis. It subsequently explores the research status of nanomaterial-based drug delivery systems for osteoporosis treatment, encompassing inorganic nanomaterials, organic nanomaterials, cell-mimicking carriers and exosomes mimics and emerging therapies targeting the osteoporosis microenvironment. Finally, the review discusses the potential of nanomedicine in treating osteoporosis and outlines the future trajectory of this burgeoning field. The aim is to provide a comprehensive reference for the application of nanomaterial-based drug delivery strategies in osteoporosis therapy, thereby fostering further advancements and innovations in this critical area of medical research.
Collapse
Affiliation(s)
- Donghong Shi
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Yuling Li
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Meng Tian
- Hebei Tourism College, Hebei, Chengde, 067000, P. R. China
| | - Mengge Xue
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| |
Collapse
|
5
|
Zhao W, Qian J, Li J, Su T, Deng X, Fu Y, Liang X, Cui H. From death to birth: how osteocyte death promotes osteoclast formation. Front Immunol 2025; 16:1551542. [PMID: 40165960 PMCID: PMC11955613 DOI: 10.3389/fimmu.2025.1551542] [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: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Bone remodeling is a dynamic and continuous process involving three components: bone formation mediated by osteoblasts, bone resorption mediated by osteoclasts, and bone formation-resorption balancing regulated by osteocytes. Excessive osteocyte death is found in various bone diseases, such as postmenopausal osteoporosis (PMOP), and osteoclasts are found increased and activated at osteocyte death sites. Currently, apart from apoptosis and necrosis as previously established, more forms of cell death are reported, including necroptosis, ferroptosis and pyroptosis. These forms of cell death play important role in the development of inflammatory diseases and bone diseases. Increasing studies have revealed that various forms of osteocyte death promote osteoclast formation via different mechanism, including actively secreting pro-inflammatory and pro-osteoclastogenic cytokines, such as tumor necrosis factor alpha (TNF-α) and receptor activator of nuclear factor-kappa B ligand (RANKL), or passively releasing pro-inflammatory damage associated molecule patterns (DAMPs), such as high mobility group box 1 (HMGB1). This review summarizes the established and potential mechanisms by which various forms of osteocyte death regulate osteoclast formation, aiming to provide better understanding of bone disease development and therapeutic target.
Collapse
Affiliation(s)
- Weijie Zhao
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jiale Qian
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Ji Li
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Tian Su
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, College of pharmacy, Hainan Medical University, Haikou, China
| | - Xiaozhong Deng
- Department of Pain Treatment, Nanxi Shan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Yonghua Fu
- Department of Hand and Foot Microsurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xuelong Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongwang Cui
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
Shariati K, Bedar M, Huang KX, Moghadam S, Mirzaie S, LaGuardia JS, Chen W, Kang Y, Ren X, Lee JC. Biomaterial Cues for Regulation of Osteoclast Differentiation and Function in Bone Regeneration. ADVANCED THERAPEUTICS 2025; 8:2400296. [PMID: 39867107 PMCID: PMC11756815 DOI: 10.1002/adtp.202400296] [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: 07/07/2024] [Indexed: 01/28/2025]
Abstract
Tissue regeneration involves dynamic dialogue between and among different cells and their surrounding matrices. Bone regeneration is specifically governed by reciprocity between osteoblasts and osteoclasts within the bone microenvironment. Osteoclast-directed resorption and osteoblast-directed formation of bone are essential to bone remodeling, and the crosstalk between these cells is vital to curating a sequence of events that culminate in the creation of bone tissue. Among bone biomaterial strategies, many have investigated the use of different material cues to direct the development and activity of osteoblasts. However, less attention has been given to exploring features that similarly target osteoclast formation and activity, with even fewer strategies demonstrating or integrating biomaterial-directed modulation of osteoblast-osteoclast coupling. This review aims to describe various biomaterial cues demonstrated to influence osteoclastogenesis and osteoclast function, emphasizing those that enhance a material construct's ability to achieve bone healing and regeneration. Additionally discussed are approaches that influence the communication between osteoclasts and osteoblasts, particularly in a manner that takes advantage of their coupling. Deepening our understanding of how biomaterial cues may dictate osteoclast differentiation, function, and influence on the microenvironment may enable the realization of bone-replacement interventions with enhanced integrative and regenerative capacities.
Collapse
Affiliation(s)
- Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Sarah Mirzaie
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
7
|
Xie W, Wan WT, Liu SY, Wang JQ, Chen C, Sun X, Liu XY, Yang Q. Causal effects of the RANK-RANKL-OPG system and scoliosis: A bidirectional 2-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40934. [PMID: 39686424 PMCID: PMC11651458 DOI: 10.1097/md.0000000000040934] [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: 08/10/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Epidemiological studies and a recent Mendelian randomization (MR) study have identified an association between low bone mass and an increased risk of scoliosis. Previous research suggests that bone loss in patients with scoliosis may be related to the RANK-RANKL-OPG system. This study is to investigate whether a causal relationship exists between the RANK-RANKL-OPG system and the development of scoliosis. Genome-wide association study (GWAS) data for RANK and RANKL were sourced from the UK Biobank's Pharmaceutical Proteomics Project, while OPG data were derived from 2 independent cohorts, and scoliosis data from the FinnGen R10 database. A bidirectional 2-sample MR framework was applied to investigate causal relationships between OPG, RANK, RANKL, and scoliosis, with inverse variance weighting (IVW) as the main analytical method. Meta-analysis was used to integrate findings across cohorts, and multiple sensitivity analyses were conducted to assess the robustness and reliability of the results. According to the IVW results, there was no significant causal relationship between RANK (OR = 0.973, 95% CI = 0.871-1.087, P = .626) and RANKL (OR = 1.048, 95% CI = 0.938-1.171, P = .411) and scoliosis. OPG is a potential protective factor for scoliosis (Folkersen 2020 OR = 0.739, 95% CI = 0.611-0.893, P = .002; Zhao 2023 OR = 0.833, 95% CI = 0.716-0.968, P = .017).The results of Meta-analysis also showed OPG (P = 1.428e-4) would reduce the risk of scoliosis. Inverse MR analysis showed no statistically significant causal relationship between scoliosis and RANK, RANKL and OPG levels (P > .05). Our study employing MR methodology provides robust evidence supporting a causal relationship between decreased osteoprotegerin (OPG) levels and increased susceptibility to scoliosis. However, no significant relationship was found between scoliosis with the RANK-RANKL-OPG system. This research establishes a basis for further exploration of the pathophysiological mechanisms and potential targeted treatments for scoliosis. Future studies are necessary to understand how OPG influences the development of scoliosis.
Collapse
Affiliation(s)
- Wei Xie
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Wen-Tao Wan
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Shuai-Yi Liu
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Jia-Qi Wang
- The First Clinical Medical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Chao Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xin-Yu Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Jinan, Shandong, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
8
|
Gostage J, Kostenuik P, Goljanek-Whysall K, Bellantuono I, McCloskey E, Bonnet N. Extra-osseous Roles of the RANK-RANKL-OPG Axis with a Focus on Skeletal Muscle. Curr Osteoporos Rep 2024; 22:632-650. [PMID: 39325366 PMCID: PMC11499344 DOI: 10.1007/s11914-024-00890-2] [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] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE OF REVIEW This review aims to consolidate recent observations regarding extra-osseous roles of the RANK-RANKL-OPG axis, primarily within skeletal muscle. RECENT FINDINGS Preclinical efforts to decipher a common signalling pathway that links the synchronous decline in bone and muscle health in ageing and disease disclosed a potential role of the RANK-RANKL-OPG axis in skeletal muscle. Evidence suggests RANKL inhibition benefits skeletal muscle function, mass, fibre-type switching, calcium homeostasis and reduces fall incidence. However, there still exists ambiguity regarding the exact mechanistic actions and subsequent functional improvements. Other potential RANK-RANKL-OPG extra-osseous roles include regulation of neural-inflammation and glucose metabolism. Growing evidence suggests the RANK-RANKL-OPG axis may play a regulatory role in extra-osseous tissues, especially in skeletal muscle. Targeting RANKL may be a novel therapy in ameliorating loss of muscle mass and function. More research is warranted to determine the causality of the RANK-RANKL-OPG axis in extra-osseous tissues, especially those affected by aging.
Collapse
Affiliation(s)
- John Gostage
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
| | - Paul Kostenuik
- School of Dentistry and Phylon Pharma Services, University of Michigan, Thousand Oaks, CA, USA
| | - Katarzyna Goljanek-Whysall
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
| | - Ilaria Bellantuono
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
| | - Eugene McCloskey
- The Medical Research Council/Versus Arthritis Centre for Integrated Research Into Musculoskeletal Aging, CIMA, University of Liverpool, Liverpool, UK
- Division of Clinical Medicine, School of Medicine and Population Health, Healthy Lifespan Institute and the Centre for Integrated Research in Musculoskeletal Aging, University of Sheffield, Sheffield, UK
| | - Nicolas Bonnet
- Service of Bone Diseases, Department of Medicine, Geneva University Hospital and Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
9
|
Kaneko K, Tsai J, Meñez D, Oh B, Suh AJ, Bae S, Mizuno M, Umemoto A, Giannopoulou E, Fujii T, Zhang Y, Stein EM, Bockman RS, Park-Min KH. Cellular signatures in human blood track bone mineral density in postmenopausal women. JCI Insight 2024; 9:e178977. [PMID: 39576015 PMCID: PMC11601907 DOI: 10.1172/jci.insight.178977] [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: 01/03/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024] Open
Abstract
Osteoclasts are the sole bone-resorbing cells and are formed by the fusion of osteoclast precursor cells (OCPs) derived from myeloid lineage cells. Animal studies reveal that circulating OCPs (cOCPs) in blood travel to bone and fuse with bone-resident osteoclasts. However, the characteristics of human cOCPs and their association with bone diseases remain elusive. We have identified and characterized human cOCPs and found a positive association between cOCPs and osteoclast activity. Sorted cOCPs have a higher osteoclastogenic potential than other myeloid cells and effectively differentiate into osteoclasts. cOCPs exhibit distinct morphology and transcriptomic signatures. The frequency of cOCPs in the blood varies among treatment-naive postmenopausal women and has an inverse correlation with lumbar spine bone density and a positive correlation with serum CTX, a bone resorption marker. The increased cOCPs in treatment-naive patients with osteoporosis were significantly diminished by denosumab, a widely used antiresorptive therapy. Our study reveals the distinctive identity of human cOCPs and the potential link between the dynamic regulation of cOCPs and osteoporosis and its treatment. Taken together, our study enhances our understanding of human cOCPs and highlights a potential opportunity to measure cOCPs through a simple blood test, which could potentially identify high-risk individuals.
Collapse
Affiliation(s)
- Kaichi Kaneko
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Division of Rheumatology, Department of Internal Medicine, Toho University Sakura Medical Center, Sakura, Chiba, Japan
| | - Jefferson Tsai
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Deniece Meñez
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Andrew Junwoo Suh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Masataka Mizuno
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Akio Umemoto
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Eugenia Giannopoulou
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Biological Sciences Department, New York City College of Technology, City University of New York, Brooklyn, New York, USA
| | - Takayuki Fujii
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Yaxia Zhang
- Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, New York, USA
- Pathology and Clinical Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Emily M. Stein
- Endocrine Service, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Richard S. Bockman
- Endocrine Service, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| |
Collapse
|
10
|
Furquim MAD, Hounkpe BW, Caparbo VF, Giardini HAM, Barbas CSV, Domiciano DS, Shinjo SK, Pereira RMR. Association between osteoprotegerin and RANKL single nucleotide polymorphisms and destructive rhinosinusitis in patients with granulomatosis with polyangiitis. BMC Rheumatol 2024; 8:63. [PMID: 39568080 PMCID: PMC11577902 DOI: 10.1186/s41927-024-00434-2] [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: 06/19/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Chronic invasive rhinosinusitis with facial bone damage is a common cause of functional and social impairment in granulomatosis with polyangiitis (GPA) patients. To the best of our knowledge, there is no clinical or laboratory biomarker to predict bone damage. METHODS This case-control study included 90 patients with GPA and 270 health controls (HCs). Patients were categorized according to the presence of tomographic facial bone erosions. Frequency of RANKL and osteoprotegerin single nucleotide polymorphisms (SNPs), analyzed by real-time polymerase chain reaction, were compared between patients and HCs, and between patients with and without bone damage. Clinical, therapeutic, and laboratory data were analyzed. RESULTS Facial bone erosion was observed in 55.5% of patients. No difference was found in the frequency of SNPs between patients with GPA and HCs. GPA patients were compared according to the presence or absence of bone damage, and a difference was found in the frequencies of osteoprotegerin G1181C (rs2073618) and RANKL A290G (rs2277438). A multivariate analysis showed that the CC genotype of osteoprotegerin 1181 was independently associated with bone erosion (OR = 3.95, CI95%=1.20-13.00, P = 0.02), as were the presence of the G allele in RANKL A290G (OR = 6.13, CI95%=1.95-19.26, P = 0.002) and higher disease duration (OR = 1.08, CI95%=1,01-1.15, P = 0.04). CONCLUSION SNPs in osteoprotegerin G1181C and RANKL A290G may play a role in the development of destructive rhinosinusitis in patients with GPA. Genetic assessment may be useful for identifying high-risk individuals. This observational study might work as a basis for further research to better understand this association and clinical trials using RANKL/osteoprotegerin as therapeutic targets.
Collapse
Affiliation(s)
- Marília A D Furquim
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil.
| | - Bidossessi W Hounkpe
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| | - Valéria F Caparbo
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| | - Henrique A M Giardini
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| | - Carmen S V Barbas
- Pneumology Division, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR, Brazil
| | - Diogo S Domiciano
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| | - Samuel K Shinjo
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| | - Rosa M R Pereira
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Paulo, Sao Paulo, SP, BR, Brazil
| |
Collapse
|
11
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
12
|
Devoy EJ, Jabari E, Kotsanos G, Choe RH, Fisher JP. An Exploration of the Role of Osteoclast Lineage Cells in Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39041616 DOI: 10.1089/ten.teb.2024.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Bone defects because of age, trauma, and surgery, which are exacerbated by medication side effects and common diseases such as osteoporosis, diabetes, and rheumatoid arthritis, are a problem of epidemic scale. The present clinical standard for treating these defects includes autografts and allografts. Although both treatments can promote robust regenerative outcomes, they fail to strike a desirable balance of availability, side effect profile, consistent regenerative efficacy, and affordability. This difficulty has contributed to the rise of bone tissue engineering (BTE) as a potential avenue through which enhanced bone regeneration could be delivered. BTE is founded upon a paradigm of using biomaterials, bioactive factors, osteoblast lineage cells (ObLCs), and vascularization to cue deficient bone tissue into a state of regeneration. Despite promising preclinical results, BTE has had modest success in being translated into the clinical setting. One barrier has been the simplicity of its paradigm relative to the complexity of biological bone. Therefore, this paradigm must be critically examined and expanded to better account for this complexity. One potential avenue for this is a more detailed consideration of osteoclast lineage cells (OcLCs). Although these cells ostensibly oppose ObLCs and bone regeneration through their resorptive functions, a myriad of investigations have shed light on their potential to influence bone equilibrium in more complex ways through their interactions with both ObLCs and bone matrix. Most BTE research has not systematically evaluated their influence. Yet contrary to expectations associated with the paradigm, a selection of BTE investigations has demonstrated that this influence can enhance bone regeneration in certain contexts. In addition, much work has elucidated the role of many controllable scaffold parameters in both inhibiting and stimulating the activity of OcLCs in parallel to bone regeneration. Therefore, this review aims to detail and explore the implications of OcLCs in BTE and how they can be leveraged to improve upon the existing BTE paradigm.
Collapse
Affiliation(s)
- Eoin J Devoy
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Erfan Jabari
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - George Kotsanos
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Robert H Choe
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
13
|
Cavalcanti de Araújo PH, Cezine MER, Vulczak A, Vieira LC, Matsuo FS, Remoto JM, Santos ADR, Miyabara EH, Alberici LC, Osako MK. RANKL signaling drives skeletal muscle into the oxidative profile. J Bone Miner Res 2024; 39:753-764. [PMID: 38619281 DOI: 10.1093/jbmr/zjae058] [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: 03/28/2023] [Revised: 03/13/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
The bone-muscle unit refers to the reciprocal regulation between bone and muscle by mechanical interaction and tissue communication via soluble factors. The RANKL stimulation induces mitochondrial biogenesis and increases the oxidative capacity in osteoclasts and adipocytes. RANKL may bind to the membrane bound RANK or to osteoprotegerin (OPG), a decoy receptor that inhibits RANK-RANKL activation. RANK is highly expressed in skeletal muscle, but the contribution of RANKL to healthy skeletal muscle fiber remains elusive. Here we show that RANKL stimulation in C2C12-derived myotubes induced activation of mitochondrial biogenesis pathways as detected by RNA-seq and western blot. RANKL expanded the mitochondrial reticulum, as shown by mitochondrial DNA quantification and MitoTracker staining, and boosted the spare respiratory capacity. Using MEK and MAPK inhibitors, we found that RANKL signals via ERK and p38 to induce mitochondrial biogenesis. The soleus from OPG-/- and OPG+/- mice showed higher respiratory rates compared to C57BL6/J WT mice, which correlates with high serum RANKL levels. RANKL infusion using a mini-osmotic pump in WT mice increased the number of mitochondria, boosted the respiratory rate, increased succinate dehydrogenase activity in skeletal muscle, and improved the fatigue resistance of gastrocnemius. Therefore, our findings reveal a new role of RANKL as an osteokine-like protein that impacts muscle fiber metabolism.
Collapse
Affiliation(s)
- Paulo Henrique Cavalcanti de Araújo
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| | - Maria Eduarda Ramos Cezine
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| | - Anderson Vulczak
- Department of Biomolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, São Paulo 14040-903, Brazil
| | - Luiz Carlos Vieira
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| | - Flávia Sayuri Matsuo
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| | - Júlia Maranghetti Remoto
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| | - Audrei Dos Reis Santos
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Elen Haruka Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, São Paulo 14040-903, Brazil
| | - Mariana Kiomy Osako
- Laboratory of Cell and Tissue Biology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo 14049-900, Brazil
| |
Collapse
|
14
|
Muralidharan A, Gomez GA, Kesavan C, Pourteymoor S, Larkin D, Tambunan W, Sechriest VF, Mohan S. Sex-Specific Effects of THRβ Signaling on Metabolic Responses to High Fat Diet in Mice. Endocrinology 2024; 165:bqae075. [PMID: 38935021 PMCID: PMC11237353 DOI: 10.1210/endocr/bqae075] [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: 03/19/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Thyroid hormone (TH) plays a crucial role in regulating the functions of both bone and adipose tissue. Given that TH exerts its cholesterol-lowering effects in hepatic tissue through the TH receptor-β (TRβ), we hypothesized that TRβ agonist therapy using MGL3196 (MGL) would be effective in treating increased adiposity and bone loss in response to a 12-week high-fat diet (HFD) in adult C57BL/6J mice. Transcriptional and serum profiling revealed that HFD-induced leptin promoted weight gain in both males and females, but MGL only suppressed leptin induction and weight gain in males. In vitro studies suggest that estrogen suppresses MGL activity in adipocytes, indicating that estrogen might interfere with MGL-TRβ function. Compared to systemic adiposity, HFD reduced bone mass in male but not female mice. Paradoxically, MGL treatment reversed macroscopic bone mineral density loss in appendicular bones, but micro-CT revealed that MGL exacerbated HFD-induced trabecular bone loss, and reduced bone strength. In studies on the mechanisms for HFD effects on bone, we found that HFD induced Rankl expression in male femurs that was blocked by MGL. By ex vivo assays, we found that RANKL indirectly represses osteoblast lineage allocation of osteoprogenitors by induction of inflammatory cytokines TNFα, IL-1β, and CCL2. Finally, we found that MGL functions in both systemic adiposity and bone by nongenomic TRβ signaling, as HFD-mediated phenotypes were not rescued in TRβ147F knockout mice with normal genomic but defective nongenomic TRβ signaling. Our findings demonstrate that the negative effects of HFD on body fat and bone phenotypes are impacted by MGL in a gender-specific manner.
Collapse
Affiliation(s)
- Aruljothi Muralidharan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Gustavo A Gomez
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Destiney Larkin
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - William Tambunan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - V Franklin Sechriest
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Orthopedic Surgery, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
15
|
Zhang D, Zhang J, Qi Q, Pan Y, Zeng R, Xu C, Liu X, Xu J, Gao M, Gao T, Zhang J, Shi S, Dong L. TNFSF11/TNFRSF11A Axis Amplifies HDM-Induced Airway Remodeling by Strengthening TGFβ1/STAT3 Action. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:399-421. [PMID: 39155739 PMCID: PMC11331193 DOI: 10.4168/aair.2024.16.4.399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/27/2024] [Accepted: 05/17/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE Asthma, an airway inflammatory disease, involves multiple tumor necrosis factors (TNF). TNF ligand superfamily member 11 (TNFSF11) and its known receptor, TNF receptor superfamily 11A (TNFRSF11A), has been implicated in asthma; however, the related mechanisms remain unknown. METHODS The serum and bronchial airway of patients with asthma and healthy subjects were examined. The air-liquid interface of primary human bronchial epithelial (HBE) cells, and Tnfsf11+/- mouse, Tnfrsf11a+/- mouse, and a humanized HSC-NOG-EXL mouse model were established. This study constructed short hairpin RNA (shRNA) of TNFSF11, TNFRSF11A, transforming growth factor β1 (TGFβ1), and transforming growth factor β receptor type 1 (TGFβR1) using lentivirus to further examine the ability of TNFSF11 protein. RESULTS This study was the first to uncover TNFSF11 overexpression in the airway and serum of asthmatic human subjects, and the TNFSF11 in serum was closely correlated with lung function. The TNFSF11/TNFRSF11A axis deficiency in Tnfsf11+/- or Tnfrsf11a+/- mice remarkably attenuated the house dust mite (HDM)-induced signal transducer and activator of transcription 3 (STAT3) action and remodeling protein expression. Similarly, the HDM-induced STAT3 action and remodeling protein expression in HBE cells decreased after pretreatment with TNFSF11 or TNFRSF11A shRNA. Meanwhile, the expression of the remodeling proteins induced by TNFSF11 significantly decreased after pretreatment with-stattic (inhibitor of STAT3 phosphorylation) in HBE cells. The STAT3 phosphorylation and remodeling protein expression induced by TNFSF11 obviously decreased after pretreatment with TGFβ1 or TGFβR1 shRNA in HBE cells. The above results also verified that blocking TNFSF11 with denosumab alleviated airway remodeling via the TGFβ1/STAT3 signaling in the humanized HSC-NOG-EXL mice with HDM-induced asthma. CONCLUSIONS TGFβ1/STAT3 action was closely correlated with TNFSF11/TNFRSF11A axis-mediated airway remodeling. This study presented a novel strategy that blocks the TNFSF11/TNFRSF11A axis to exert a protective effect against asthma.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jintao Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Qian Qi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Rong Zeng
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Changjuan Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Xiaofei Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jiawei Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Mingxia Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Tingting Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Shuochuan Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China.
| |
Collapse
|
16
|
Frey S, Gérard M, Guillot P, Wargny M, Bach-Ngohou K, Bigot-Corbel E, Renaud Moreau N, Caillard C, Mirallié E, Cariou B, Blanchard C. Parathyroidectomy Improves Bone Density in Women With Primary Hyperparathyroidism and Preoperative Osteopenia. J Clin Endocrinol Metab 2024; 109:1494-1504. [PMID: 38152848 DOI: 10.1210/clinem/dgad718] [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: 09/14/2023] [Indexed: 12/29/2023]
Abstract
CONTEXT Osteoporosis and/or bone fractures are indications of parathyroidectomy in primary hyperparathyroidism (PHPT), especially in women. However, the benefit of surgery in patients with osteopenia remains unclear. OBJECTIVE To evaluate bone mineral density (BMD) and bone remodeling biomarkers changes 1 year after parathyroidectomy in women with PHPT. DESIGN In the prospective, monocentric, observational prospective cohort with primary hyperparathyroidism patients (CoHPT) cohort, women operated for sporadic PHPT since 2016 with ≥1 year follow-up were included. BMD (dual-X ray absorptiometry) and bone remodeling biomarkers [cross-linked C-telopeptide (CTX), procollagen type 1 N-terminal propeptide (P1NP), and bone-specific alkaline phosphatases] were assessed before and 1 year after parathyroidectomy. SETTING Referral center. PATIENTS A total of 177 women with PHPT (62.5 ± 13.3 years, 83.1% menopausal, 43.9% osteopenic, and 45.1% osteoporotic) were included. INTERVENTION Parathyroidectomy. MAIN OUTCOME MEASURE BMD change between before and 1 year after parathyroidectomy. RESULTS Parathyroidectomy resulted in significant increase in BMD and decrease in serum bone remodeling biomarker concentrations. In the 72 patients with baseline osteopenia, mean BMD significantly increased at the lumbar spine [+0.05 g/cm2 (95% confidence interval [CI], 0.03-0.07)], the femoral neck [+0.02 g/cm2 (95% CI 0.00-0.04)], the total hip [+0.02 g/cm2 (95% CI 0.01-0.02)], and the forearm [+0.01 (95% CI 0.00-0.02)], comparable to osteoporotic patients. Among osteopenic patients, those with individual BMD gain (>0.03 g/cm2) at ≥1 site had higher preoperative serum CTX, P1NP, and urine calcium concentrations than those without improvement. CONCLUSION Parathyroidectomy significantly improved BMD and remodeling biomarkers in women with osteopenia, thereby supporting the benefit of parathyroidectomy in these patients. Preoperative serum CTX and P1NP concentrations could be useful to predict expected BMD gain.
Collapse
Affiliation(s)
- Samuel Frey
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Maxime Gérard
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
| | - Pascale Guillot
- Nantes Université, CHU Nantes, Service de Rhumatologie, F-44000 Nantes, France
| | - Matthieu Wargny
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
- Nantes Université, CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, F-44000 Nantes, France
| | - Kalyane Bach-Ngohou
- Nantes Université, CHU Nantes, Department of Biochemistry and INSERM, The Enteric Nervous System in Gut and Brain Disorders, IMAD, F-4400 Nantes, France
| | - Edith Bigot-Corbel
- Nantes Université, CHU Nantes, Laboratoire de biochimie, Hôpital Guillaume et René Laennec, F-4400 Nantes, France
| | - Nelly Renaud Moreau
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
| | - Cécile Caillard
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
| | - Eric Mirallié
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Claire Blanchard
- Nantes Université, CHU Nantes, Chirurgie Cancérologique, Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, F-44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| |
Collapse
|
17
|
Li X, Zhang C, Feng C, Zhang Z, Feng N, Sha H, Luo X, Zou G, Liang H. Transcriptome Analysis Elucidates the Potential Key Genes Involved in Rib Development in bmp6-Deficient Silver Carp ( Hypophthalmichthys molitrix). Animals (Basel) 2024; 14:1451. [PMID: 38791669 PMCID: PMC11117292 DOI: 10.3390/ani14101451] [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: 04/03/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Bone morphogenetic protein 6 (BMP-6) is a constituent of the TGF-β superfamily, known for its ability to stimulate bone and cartilage formation. The investigation of bmp6's involvement in the formation of intermuscular bones in fish has garnered significant attention in recent years. The rib cage is an important skeletal structure that plays a protective function for internal organs in fish. However, there has been limited research conducted on the effects of the bmp6 gene on rib development. Silver carp is one of four major fish in China, favoured for its affordability and tender muscle. Nevertheless, the presence of numerous intermuscular bones in silver carp significantly hinders the advancement of its palatability and suitability for processing. This study showcases the effective utilisation of CRISPR/Cas9 technology for the purpose of disrupting the bmp6 gene in silver carp, leading to the creation of chimeras in the P0 generation, marking the first instance of such an achievement. The chimeras exhibited complete viability, normal appearance, and partial intermuscular bones loss, with approximately 30% of them displaying rib bifurcation or bending. Subsequently, a transcriptome analysis on ribs of P0 chimeras and wild-type silver carp was conducted, leading to the identification of 934 genes exhibiting differential expression, of which 483 were found to be up-regulated and 451 were found to be down-regulated. The results of the KEGG analysis revealed that the "NF-kappa B signalling pathway", "Hippo signalling pathway", "osteoclast differentiation", and "haematopoietic cell lineage" exhibited enrichment and displayed a significant correlation with bone development. The up-regulated genes such as tnfα, fos, and ctgf in pathways may facilitate the proliferation and differentiation of osteoclasts, whereas the down-regulation of genes such as tgfb2 and tgfbr1 in pathways may hinder the formation and specialisation of osteoblasts, ultimately resulting in rib abnormalities. This study presents novel findings on the impact of bmp6 gene deletion on the rib development of silver carp, while simultaneously investigating the previously unexplored molecular mechanisms underlying rib defects in fish.
Collapse
Affiliation(s)
- Xiaohui Li
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Chunyan Zhang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Cui Feng
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Zewen Zhang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Nannan Feng
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Hang Sha
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Xiangzhong Luo
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Guiwei Zou
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Hongwei Liang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
18
|
Maji S, Kumar A, Emdad L, Fisher PB, Das SK. Molecular landscape of prostate cancer bone metastasis. Adv Cancer Res 2024; 161:321-365. [PMID: 39032953 DOI: 10.1016/bs.acr.2024.04.007] [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] [Indexed: 07/23/2024]
Abstract
Prostate cancer (PC) has a high propensity to develop bone metastases, causing severe pain and pathological fractures that profoundly impact a patients' normal functions. Current clinical intervention is mainly palliative focused on pain management, and tumor progression is refractory to standard therapeutic regimens. This limited treatment efficacy is at least partially due to a lack of comprehensive understanding of the molecular landscape of the disease pathology, along with the intensive overlapping of physiological and pathological molecular signaling. The niche is overwhelmed with diverse cell types with inter- and intra-heterogeneity, along with growth factor-enriched cells that are supportive of invading cell proliferation, providing an additional layer of complexity. This review seeks to provide molecular insights into mechanisms underlying PC bone metastasis development and progression.
Collapse
Affiliation(s)
- Santanu Maji
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
19
|
Wang T, Tang Y, Xia Y, Zhang Q, Cao S, Bie M, Kang F. IGF2 promotes alveolar bone regeneration in murine periodontitis via inhibiting cGAS/STING-mediated M1 macrophage polarization. Int Immunopharmacol 2024; 132:111984. [PMID: 38565043 DOI: 10.1016/j.intimp.2024.111984] [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: 01/01/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Periodontitis is a chronic inflammatory disease with the destruction of supporting periodontal tissue. This study evaluated the role of insulin-like growth factor 2 (IGF2) in periodontitis by inhibiting the polarization of M1 macrophages via the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway. IGF2 was enriched in the gingival tissue of murine periodontitis model identified by RNA sequencing. IGF2 application alleviated the expression of pro-inflammatory factors and promoted osteogenesis and the expression of related genes and proteins in a dose-dependent manner in periodontitis. The result of micro-CT verified this finding. Both in vivo and in vitro results revealed that IGF2 decreased the polarization of M1 macrophages and pro-inflammatory factors by immunofluorescence staining, flow cytometry, western blotting and RT-PCR. IGF2 application promoted the osteogenic ability of periodontal ligament fibroblasts (PDLFs) indirectly via its inhibition of M1 polarization evaluated by alkaline phosphatase and alizarin red staining. Then, the cGAS/STING pathway was upregulated in periodontitis and macrophages challenged by LPS, the inhibition of which led to downregulation of M1 polarization. Furthermore, IGF2 could downregulate cGAS, STING and the phosphorylation of P65. Collectively, our study indicates IGF2 can regulate the polarization of M1 macrophages via the cGAS/STING pathway and highlights the promising future of IGF2 as a therapeutic treatment for periodontitis.
Collapse
Affiliation(s)
- Tairan Wang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yi Tang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yuxing Xia
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Qian Zhang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Shaokang Cao
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Miaomiao Bie
- Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feiwu Kang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
20
|
Kim JE, Park SG, Ka DB, Kim EK, Cho SM, Kim HR, Lee MN, Choi KC, Yoon WK, Nam KH. Phf7 has impacts on the body growth and bone remodeling by regulating testicular hormones in male mice. Biochem Biophys Res Commun 2024; 704:149596. [PMID: 38430697 DOI: 10.1016/j.bbrc.2024.149596] [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: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 03/05/2024]
Abstract
PHD finger protein 7 (Phf7) is a member of the PHF family proteins, which plays important roles in spermiogenesis. Phf7 is expressed in the adult testes and its deficiency causes male infertility. In this study, we tried to find the causal relationship between Phf7 deficiency and reduced growth retardation which were found in null knock-out (Phf7-/-) mice. Phf7-/- mice were born normally in the Mendelian ratio. However, the Phf7-/- males showed decreased body weight gain, bone mineral density, and bone mineral content compared to those in wild-type (WT) mice. Histological analysis for tibia revealed increased number of osteoclast cells in Phf7-/- mice compared with that in WT mice. When we analyzed the expressions for marker genes for the initial stage of osteoclastogenesis, such as receptor activator of nuclear factor kappa B (Rank) in tibia, there was no difference in the mRNA levels between Phf7-/- and WT mice. However, the expression of tartrate-resistant acid phosphatase (Trap), a mature stage marker gene, was significantly higher in Phf7-/- mice than in WT mice. In addition, the levels of testosterone and dihydrotestosterone (DHT), more potent and active form of testosterone, were significantly reduced in the testes of Phf7-/- mice compared to those in WT mice. Furthermore, testicular mRNA levels for steroidogenesis marker genes, namely Star, Cyp11a1, Cyp17a1 and 17β-hsd, were significantly lower in Phf7-/- mice than in WT mice. In conclusion, these results suggest that Phf7 deficiency reduces the production of male sex hormones and thereby impairs associated bone remodeling.
Collapse
Affiliation(s)
- Ji Eun Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Seul Gi Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Dan Bi Ka
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Eun-Kyoung Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Sang-Mi Cho
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Hae-Rim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Mi Ni Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Kyung-Chul Choi
- College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Won Kee Yoon
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Ki-Hoan Nam
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| |
Collapse
|
21
|
Giorello MB, Borzone FR, Mora MF, Padin MDR, Wernicke A, Labovsky V, Chasseing NA. RANK in cancer-associated fibroblasts: A valuable prognostic determinant for metastasis in early-stage breast cancer patients. Cancer Biomark 2024; 41:115-132. [PMID: 39240628 PMCID: PMC11492045 DOI: 10.3233/cbm-230523] [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: 01/10/2024] [Accepted: 07/19/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND The molecular system of receptor activator of nuclear factor kappa-β (RANK) and its ligand (RANKL) plays a role in a variety of physiological and pathological processes. These encompass the regulation of bone metabolism, mammary gland development, immune function, as well as their involvement and tumorigenesis. Nevertheless, limited knowledge exists regarding their function within the tumor microenvironment. METHODS AND RESULTS We explored the significance of RANK expression in cancer-associated fibroblasts (CAFs) as a prognostic biomarker in early breast cancer patients (BCPs) by immunohistochemistry. Results reveal a significant correlation between high RANK expression in CAFs and an increased risk of metastasis (p= 0.006), shorter metastasis-free survival (MFS) [p= 0.007, OR (95%CI) = 2.290 (1.259-4.156)], and lower overall survival (OS) [p= 0.004, OR (95%CI) = 2.469 (1.343-4.541)]. Upon analyzing the phenotype of CD34(-) CAFs isolated from primary tumors in BCPs, we observed co-expression of RANK with CD105 marker by immunofluorescence and flow cytometry, characteristic of mesenchymal stem/stromal cells (MSCs), suggesting the possible cellular origin. Also RANKL-RANK system increase the OCT-4, SOX-2 and DKK-1 (dickkopf 1) gene expression in CD34(-) CAFs by RT-PCR. Moreover, this system plays a crucial role in the migration of these CD34(-) CAFs. CONCLUSIONS These results support the clinical relevance of RANK in CAFs and propose its potential as a future therapeutic target in the treatment of early BCPs.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María Florencia Mora
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María del Rosario Padin
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Wernicke
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Departamento de Anatomía Patológica, Hospital Italiano, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Pius AK, Toya M, Gao Q, Lee ML, Ergul YS, Chow SKH, Goodman SB. Effects of Aging on Osteosynthesis at Bone-Implant Interfaces. Biomolecules 2023; 14:52. [PMID: 38254652 PMCID: PMC10813487 DOI: 10.3390/biom14010052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Joint replacement is a common surgery and is predominantly utilized for treatment of osteoarthritis in the aging population. The longevity of many of these implants depends on bony ingrowth. Here, we provide an overview of current techniques in osteogenesis (inducing bone growth onto an implant), which is affected by aging and inflammation. In this review we cover the biologic underpinnings of these processes as well as the clinical applications. Overall, aging has a significant effect at the cellular and macroscopic level that impacts osteosynthesis at bone-metal interfaces after joint arthroplasty; potential solutions include targeting prolonged inflammation, preventing microbial adhesion, and enhancing osteoinductive and osteoconductive properties.
Collapse
Affiliation(s)
- Alexa K. Pius
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Masakazu Toya
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Qi Gao
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Max L. Lee
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Yasemin Sude Ergul
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
| | - Stuart Barry Goodman
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94063, USA; (A.K.P.); (M.T.); (Q.G.); (M.L.L.); (Y.S.E.); (S.K.-H.C.)
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
24
|
Padovano C, Bianco SD, Sansico F, De Santis E, Tamiro F, Colucci M, Totti B, Di Iasio S, Bruno G, Panelli P, Miscio G, Mazza T, Giambra V. The Notch1 signaling pathway directly modulates the human RANKL-induced osteoclastogenesis. Sci Rep 2023; 13:21199. [PMID: 38040752 PMCID: PMC10692129 DOI: 10.1038/s41598-023-48615-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023] Open
Abstract
Notch signaling is an evolutionary conserved pathway with a key role in tissue homeostasis, differentiation and proliferation. It was reported that Notch1 receptor negatively regulates mouse osteoclast development and formation by inhibiting the expression of macrophage colony-stimulating factor in mesenchymal cells. Nonetheless, the involvement of Notch1 pathway in the generation of human osteoclasts is still controversial. Here, we report that the constitutive activation of Notch1 signaling induced a differentiation block in human mononuclear CD14+ cells directly isolated from peripheral blood mononuclear cells (PBMCs) upon in vitro stimulation to osteoclasts. Additionally, using a combined approach of single-cell RNA sequencing (scRNA-Seq) simultaneously with a panel of 31 oligo-conjugated antibodies against cell surface markers (AbSeq assay) as well as unsupervised learning methods, we detected four different cell stages of human RANKL-induced osteoclastogenesis after 5 days in which Notch1 signaling enforces the cell expansion of specific subsets. These cell populations were characterized by distinct gene expression and immunophenotypic profiles and active Notch1, JAK/STAT and WNT signaling pathways. Furthermore, cell-cell communication analyses revealed extrinsic modulators of osteoclast progenitors including the IL7/IL7R and WNT5a/RYK axes. Interestingly, we also report that Interleukin-7 receptor (IL7R) was a downstream effector of Notch1 pathway and that Notch1 and IL7R interplay promoted cell expansion of human RANKL-induced osteoclast progenitors. Taken together, these findings underline a novel cell pattern of human osteoclastogenesis, outlining the key role of Notch1 and IL-7R signaling pathways.
Collapse
Affiliation(s)
- Costanzo Padovano
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Salvatore Daniele Bianco
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
| | - Francesca Sansico
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Elisabetta De Santis
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Francesco Tamiro
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Mattia Colucci
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Beatrice Totti
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Serena Di Iasio
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Gaja Bruno
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Patrizio Panelli
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Giuseppe Miscio
- Clinical Laboratory Analysis and Transfusional Medicine, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
| | - Vincenzo Giambra
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy.
| |
Collapse
|
25
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
26
|
Takami K, Okamoto K, Etani Y, Hirao M, Miyama A, Okamura G, Goshima A, Miura T, Kurihara T, Fukuda Y, Kanamoto T, Nakata K, Okada S, Ebina K. Anti-NF-κB peptide derived from nuclear acidic protein attenuates ovariectomy-induced osteoporosis in mice. JCI Insight 2023; 8:e171962. [PMID: 37991021 PMCID: PMC10721323 DOI: 10.1172/jci.insight.171962] [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: 05/03/2023] [Accepted: 10/10/2023] [Indexed: 11/23/2023] Open
Abstract
NF-κB is a transcription factor that is activated with aging. It plays a key role in the development of osteoporosis by promoting osteoclast differentiation and inhibiting osteoblast differentiation. In this study, we developed a small anti-NF-κB peptide called 6A-8R from a nuclear acidic protein (also known as macromolecular translocation inhibitor II, Zn2+-binding protein, or parathymosin) that inhibits transcriptional activity of NF-κB without altering its nuclear translocation and binding to DNA. Intraperitoneal injection of 6A-8R attenuated ovariectomy-induced osteoporosis in mice by inhibiting osteoclast differentiation, promoting osteoblast differentiation, and inhibiting sclerostin production by osteocytes in vivo with no apparent side effects. Conversely, in vitro, 6A-8R inhibited osteoclast differentiation by inhibiting NF-κB transcriptional activity, promoted osteoblast differentiation by promoting Smad1 phosphorylation, and inhibited sclerostin expression in osteocytes by inhibiting myocyte enhancer factors 2C and 2D. These findings suggest that 6A-8R has the potential to be an antiosteoporotic therapeutic agent with uncoupling properties.
Collapse
Affiliation(s)
- Kenji Takami
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Orthopaedic Surgery, Nippon Life Hospital, Nishi-ku, Osaka, Japan
| | - Kazuki Okamoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuki Etani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Makoto Hirao
- Department of Orthopaedic Surgery, National Hospital Organization Osaka Minami Medical Center, Kawachinagano, Osaka, Japan
| | - Akira Miyama
- Department of Orthopaedic Surgery, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Osaka, Japan
| | - Gensuke Okamura
- Department of Orthopaedic Surgery, Osaka Rosai Hospital, Kita-ku, Sakai, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Taihei Miura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takuya Kurihara
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuji Fukuda
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Ken Nakata
- Department of Health and Sport Sciences, and
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kosuke Ebina
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
27
|
Cho SH, Lee S, Park JI, La Yang Y, Kim SR, Ahn J, Jeong H, Jung HY, Gwak N, Kim KN, Kim Y. Age-associated spinal stenosis in the turquoise killifish. iScience 2023; 26:107877. [PMID: 37810235 PMCID: PMC10550727 DOI: 10.1016/j.isci.2023.107877] [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: 09/28/2022] [Revised: 03/22/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Aging triggers spinal degeneration, including common spinal stenosis, which causes back and leg pain in older individuals, significantly impacting their quality of life. Here, we explored aging traits in turquoise killifish spines, potentially offering a model for age-linked spinal stenosis in humans. Aged turquoise killifish exhibited body shape deformation and increased vertebral collapse, which was further accelerated by spawning. High-resolution CT scans revealed suppressed cortical bone thickness and hemal arch area in vertebrae due to spawning, and osteophyte formation was observed in both aged and breeding fish populations. Scale mineralization mirrored these changes, increasing with age but being suppressed by spawning. The expression of sp7, sox9b, axin1, and wnt4a/b genes can be utilized to monitor age- and reproduction-dependent spine deformation. This study demonstrates that turquoise killifish and humans share certain phenotypes of age-related vertebral abnormalities, suggesting that turquoise killifish could serve as a potential model for studying human spinal stenosis.
Collapse
Affiliation(s)
- Su-Hyeon Cho
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
- Department of Medical Biomaterials Engineering, College of Biomedical Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seongsin Lee
- Center for Plant Aging Research, Institute for Basic Science, Daegu 42988, Republic of Korea
| | - Jae-Il Park
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Yoon La Yang
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Song-Rae Kim
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
- Department of Medical Biomaterials Engineering, College of Biomedical Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Juhee Ahn
- Department of Medical Biomaterials Engineering, College of Biomedical Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hoibin Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
- Seoul Center, Korea Basic Science Institute, Seoul 02841, Republic of Korea
| | - Hye-Yeon Jung
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Nayoung Gwak
- Center for Plant Aging Research, Institute for Basic Science, Daegu 42988, Republic of Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
- Department of Bio-analysis Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Yumi Kim
- Center for Plant Aging Research, Institute for Basic Science, Daegu 42988, Republic of Korea
- Center for Genome Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| |
Collapse
|
28
|
Giannoni P, Marini C, Cutrona G, Sambuceti GM, Fais F, de Totero D. Unraveling the Bone Tissue Microenvironment in Chronic Lymphocytic Leukemia. Cancers (Basel) 2023; 15:5058. [PMID: 37894425 PMCID: PMC10605026 DOI: 10.3390/cancers15205058] [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: 09/14/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most frequent leukemia in Western countries. Although characterized by the progressive expansion and accumulation of leukemic B cells in peripheral blood, CLL cells develop in protective niches mainly located within lymph nodes and bone marrow. Multiple interactions between CLL and microenvironmental cells may favor the expansion of a B cell clone, further driving immune cells toward an immunosuppressive phenotype. Here, we summarize the current understanding of bone tissue alterations in CLL patients, further addressing and suggesting how the multiple interactions between CLL cells and osteoblasts/osteoclasts can be involved in these processes. Recent findings proposing the disruption of the endosteal niche by the expansion of a leukemic B cell clone appear to be a novel field of research to be deeply investigated and potentially relevant to provide new therapeutic approaches.
Collapse
Affiliation(s)
- Paolo Giannoni
- Department of Experimental Medicine, Biology Section, University of Genova, 16132 Genova, Italy;
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (G.M.S.)
- CNR Institute of Bioimages and Molecular Physiology, 20054 Milano, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (G.C.); (F.F.)
| | - Gian Mario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (G.M.S.)
- Department of Health Sciences, University of Genova, 16132 Genova, Italy
| | - Franco Fais
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (G.C.); (F.F.)
- Department of Experimental Medicine, Anatomy Section, University of Genova, 16132 Genova, Italy
| | - Daniela de Totero
- Department of Health Sciences, University of Genova, 16132 Genova, Italy
| |
Collapse
|
29
|
Lee SH, Park SY, Kim JH, Kim N, Lee J. Ginsenoside Rg2 inhibits osteoclastogenesis by downregulating the NFATc1, c-Fos, and MAPK pathways. BMB Rep 2023; 56:551-556. [PMID: 37605614 PMCID: PMC10618073 DOI: 10.5483/bmbrep.2023-0100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 05/25/2025] Open
Abstract
Ginsenosides, among the most active components of ginseng, exhibit several therapeutic effects against cancer, diabetes, and other metabolic diseases. However, the molecular mechanism underlying the anti-osteoporotic activity of ginsenoside Rg2, a major ginsenoside, has not been clearly elucidated. This study aimed to determine the effects of ginsenoside Rg2 on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation. Results indicate that ginsenoside Rg2 inhibits RANKLinduced osteoclast differentiation of bone marrow macrophages (BMMs) without cytotoxicity. Pretreatment with ginsenoside Rg2 significantly reduced the RANKL-induced gene expression of c-fos and nuclear factor of activated T-cells (Nfatc1), as well as osteoclast-specific markers tartrate-resistant acid phosphatase (TRAP, Acp5) and osteoclast-associated receptor (Oscar). Moreover, RANKL-induced phosphorylation of mitogen-activated protein kinases (MAPKs) was decreased by ginsenoside Rg2 in BMM. Therefore, we suggest that ginsenoside Rg2 suppresses RANKLinduced osteoclast differentiation through the regulation of MAPK signaling-mediated osteoclast markers and could be developed as a therapeutic drug for the prevention and treatment of osteoporosis. [BMB Reports 2023; 56(10): 551-556].
Collapse
Affiliation(s)
- Sung-Hoon Lee
- Department of Life Science and Genetic Engineering, Graduate School of PaiChai University, Daejeon 35345, Korea
| | - Shin-Young Park
- Division of Software Engineering, PaiChai University, Daejeon 35345, Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Junwon Lee
- Department of Life Science and Genetic Engineering, Graduate School of PaiChai University, Daejeon 35345, Korea
| |
Collapse
|
30
|
Lee SH, Park SY, Kim JH, Kim N, Lee J. Ginsenoside Rg2 inhibits osteoclastogenesis by downregulating the NFATc1, c-Fos, and MAPK pathways. BMB Rep 2023; 56:551-556. [PMID: 37605614 PMCID: PMC10618073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
Ginsenosides, among the most active components of ginseng, exhibit several therapeutic effects against cancer, diabetes, and other metabolic diseases. However, the molecular mechanism underlying the anti-osteoporotic activity of ginsenoside Rg2, a major ginsenoside, has not been clearly elucidated. This study aimed to determine the effects of ginsenoside Rg2 on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation. Results indicate that ginsenoside Rg2 inhibits RANKLinduced osteoclast differentiation of bone marrow macrophages (BMMs) without cytotoxicity. Pretreatment with ginsenoside Rg2 significantly reduced the RANKL-induced gene expression of c-fos and nuclear factor of activated T-cells (Nfatc1), as well as osteoclast-specific markers tartrate-resistant acid phosphatase (TRAP, Acp5) and osteoclast-associated receptor (Oscar). Moreover, RANKL-induced phosphorylation of mitogen-activated protein kinases (MAPKs) was decreased by ginsenoside Rg2 in BMM. Therefore, we suggest that ginsenoside Rg2 suppresses RANKLinduced osteoclast differentiation through the regulation of MAPK signaling-mediated osteoclast markers and could be developed as a therapeutic drug for the prevention and treatment of osteoporosis. [BMB Reports 2023; 56(10): 551-556].
Collapse
Affiliation(s)
- Sung-Hoon Lee
- Department of Life Science and Genetic Engineering, Graduate School of PaiChai University, Daejeon 35345, Korea
| | - Shin-Young Park
- Division of Software Engineering, PaiChai University, Daejeon 35345, Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Junwon Lee
- Department of Life Science and Genetic Engineering, Graduate School of PaiChai University, Daejeon 35345, Korea
| |
Collapse
|
31
|
Boyce BF, Li J, Yao Z, Xing L. Nuclear Factor-Kappa B Regulation of Osteoclastogenesis and Osteoblastogenesis. Endocrinol Metab (Seoul) 2023; 38:504-521. [PMID: 37749800 PMCID: PMC10613774 DOI: 10.3803/enm.2023.501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 09/27/2023] Open
Abstract
Maintenance of skeletal integrity requires the coordinated activity of multinucleated bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts form resorption lacunae on bone surfaces in response to cytokines by fusion of precursor cells. Osteoblasts are derived from mesenchymal precursors and lay down new bone in resorption lacunae during bone remodeling. Nuclear factorkappa B (NF-κB) signaling regulates osteoclast and osteoblast formation and is activated in osteoclast precursors in response to the essential osteoclastogenic cytokine, receptor activator of NF-κB ligand (RANKL), which can also control osteoblast formation through RANK-RANKL reverse signaling in osteoblast precursors. RANKL and some pro-inflammatory cytokines, including tumor necrosis factor (TNF), activate NF-κB signaling to positively regulate osteoclast formation and functions. However, these cytokines also limit osteoclast and osteoblast formation through NF-κB signaling molecules, including TNF receptor-associated factors (TRAFs). TRAF6 mediates RANKL-induced osteoclast formation through canonical NF-κB signaling. In contrast, TRAF3 limits RANKL- and TNF-induced osteoclast formation, and it restricts transforming growth factor β (TGFβ)-induced inhibition of osteoblast formation in young and adult mice. During aging, neutrophils expressing TGFβ and C-C chemokine receptor type 5 (CCR5) increase in bone marrow of mice in response to increased NF-κB-induced CC motif chemokine ligand 5 (CCL5) expression by mesenchymal progenitor cells and injection of these neutrophils into young mice decreased bone mass. TGFβ causes degradation of TRAF3, resulting in decreased glycogen synthase kinase-3β/β-catenin-mediated osteoblast formation and age-related osteoporosis in mice. The CCR5 inhibitor, maraviroc, prevented accumulation of TGFβ+/CCR5+ neutrophils in bone marrow and increased bone mass by inhibiting bone resorption and increasing bone formation in aged mice. This paper updates current understanding of how NF-κB signaling is involved in the positive and negative regulation of cytokine-mediated osteoclast and osteoblast formation and activation with a focus on the role of TRAF3 signaling, which can be targeted therapeutically to enhance bone mass.
Collapse
Affiliation(s)
- Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Jinbo Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
32
|
Ma C, Li X, Xiao H, Li B, Gu H, Guo Y, Wang H, Wen Y, Chen L. Course-, dose-, and stage-dependent toxic effects of prenatal acetaminophen exposure on fetal long bone development. Toxicol Lett 2023; 387:50-62. [PMID: 37741353 DOI: 10.1016/j.toxlet.2023.09.007] [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: 04/28/2023] [Revised: 08/24/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Abstract
Acetaminophen is a common analgesic and fever reduction medicine for pregnant women. Epidemiological studies suggest that prenatal acetaminophen exposure (PAcE) affects offspring health and development. However, the effects of PAcE on fetal long bone development and its potential mechanisms have not been elucidated. Based on clinical dosing characteristics, fetal mouse femurs were obtained for detection after oral gavage of acetaminophen at different doses (0, 100 or 400 mg/kg d), courses (single or multiple times) or stages (mid- or late pregnancy) during pregnancy in Kunming mice. The results showed that compared with the control group, PAcE reduced the length of total femur and the primary ossification center (POC), delayed the mineralization of POC and the ossification of epiphyseal region, and down-regulated the mRNA expression of osteogenic function markers (such as Runx2, Bsp, Ocn , Col1a1) in fetal femur, particularly in the high dose, multiple courses, and mid-pregnancy group. Meanwhile, the osteoclast and angiogenic function were also inhibited by PAcE at high dose, multiple courses, and mid-pregnancy, but the inhibition level was less than osteogenic function. Moreover, the alteration of canonical Wnt signalling pathway in PAcE fetal bone were consistent with its osteogenesis function changes. In conclusion, PAcE caused development toxicity and multi-cellular function inhibition in fetal long bone, particularly in the high dose, multiple treatments and mid-pregnancy group, and the alteration of canonical Wnt signalling pathway may be its potential mechanism.
Collapse
Affiliation(s)
- Chi Ma
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xufeng Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Joint Disease Research Center of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Joint Disease Research Center of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hanwen Gu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Joint Disease Research Center of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Joint Disease Research Center of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
33
|
Stein M, Elefteriou F, Busse B, Fiedler IA, Kwon RY, Farell E, Ahmad M, Ignatius A, Grover L, Geris L, Tuckermann J. Why Animal Experiments Are Still Indispensable in Bone Research: A Statement by the European Calcified Tissue Society. J Bone Miner Res 2023; 38:1045-1061. [PMID: 37314012 PMCID: PMC10962000 DOI: 10.1002/jbmr.4868] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 05/03/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
Major achievements in bone research have always relied on animal models and in vitro systems derived from patient and animal material. However, the use of animals in research has drawn intense ethical debate and the complete abolition of animal experimentation is demanded by fractions of the population. This phenomenon is enhanced by the reproducibility crisis in science and the advance of in vitro and in silico techniques. 3D culture, organ-on-a-chip, and computer models have improved enormously over the last few years. Nevertheless, the overall complexity of bone tissue cross-talk and the systemic and local regulation of bone physiology can often only be addressed in entire vertebrates. Powerful genetic methods such as conditional mutagenesis, lineage tracing, and modeling of the diseases enhanced the understanding of the entire skeletal system. In this review endorsed by the European Calcified Tissue Society (ECTS), a working group of investigators from Europe and the US provides an overview of the strengths and limitations of experimental animal models, including rodents, fish, and large animals, as well the potential and shortcomings of in vitro and in silico technologies in skeletal research. We propose that the proper combination of the right animal model for a specific hypothesis and state-of-the-art in vitro and/or in silico technology is essential to solving remaining important questions in bone research. This is crucial for executing most efficiently the 3R principles to reduce, refine, and replace animal experimentation, for enhancing our knowledge of skeletal biology, and for the treatment of bone diseases that affect a large part of society. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Merle Stein
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), University Medical Center Hamburg-Eppendorf, Germany
| | - Imke A.K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), University Medical Center Hamburg-Eppendorf, Germany
| | - Ronald Young Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, USA and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Eric Farell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Mubashir Ahmad
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Liam Grover
- Healthcare Technologies Institute, Institute of Translational MedicineHeritage Building Edgbaston, Birmingham
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, Liège, Belgium
- Skeletal Biology & Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| |
Collapse
|
34
|
Lin S, Li M, Zhou Y, Chen L, Wang Y, Zhuang Z, Zhao H, Yang R. Annexin A3 accelerates osteoclast differentiation by promoting the level of RANK and TRAF6. Bone 2023; 172:116758. [PMID: 37030499 DOI: 10.1016/j.bone.2023.116758] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 04/10/2023]
Abstract
Annexin A3 (ANXA3), a member of Annexin family, is reported to mediate membrane transport and cancer development. However, the effect of ANXA3 on osteoclast formation and bone metabolism is still unclear. In this study, we found that knockdown of ANXA3 can significantly inhibit receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation through NF-κB signaling. ANXA3 downregulation abrogated the expression of osteoclast-specific genes, including Acp5, Mmp9 and Ctsk in osteoclast precursors. Moreover, lentiviral of shRNA against ANXA3 reversed the bone loss in osteoporosis using ovariectomized mice model. Mechanistically, we found that ANXA3 directly bound to RANK and TRAF6 to accelerate osteoclast differentiation by promoting their transcription and limiting degradation. In conclusion, we propose a fundamentally novel RANK-ANXA3-TRAF6 complex to effectively modulate the formation and differentiation of osteoclast to manipulate bone metabolism. The ANXA3-targeted therapeutic strategy may provide new insight for bone degrading-related diseases prevention and treatment.
Collapse
Affiliation(s)
- Shuai Lin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China; Chinese Institute for Brain Research, Changping District, Beijing, China
| | - Mingzhao Li
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China; Chinese Institute for Brain Research, Changping District, Beijing, China
| | - Yikun Zhou
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Liujing Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yiming Wang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zimeng Zhuang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Hu Zhao
- Chinese Institute for Brain Research, Changping District, Beijing, China.
| | - Ruili Yang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Haidian District, Beijing, China; National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China; Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China.
| |
Collapse
|
35
|
Ma Y, Xu Y, Zhang Y, Duan X. Molecular Mechanisms of Craniofacial and Dental Abnormalities in Osteopetrosis. Int J Mol Sci 2023; 24:10412. [PMID: 37373559 DOI: 10.3390/ijms241210412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Osteopetrosis is a group of genetic bone disorders characterized by increased bone density and defective bone resorption. Osteopetrosis presents a series of clinical manifestations, including craniofacial deformities and dental problems. However, few previous reports have focused on the features of craniofacial and dental problems in osteopetrosis. In this review, we go through the clinical features, types, and related pathogenic genes of osteopetrosis. Then we summarize and describe the characteristics of craniofacial and dental abnormalities in osteopetrosis that have been published in PubMed from 1965 to the present. We found that all 13 types of osteopetrosis have craniomaxillofacial and dental phenotypes. The main pathogenic genes, such as chloride channel 7 gene (CLCN7), T cell immune regulator 1 (TCIRG1), osteopetrosis-associated transmembrane protein 1 (OSTM1), pleckstrin homology domain-containing protein family member 1 (PLEKHM1), and carbonic anhydrase II (CA2), and their molecular mechanisms involved in craniofacial and dental phenotypes, are discussed. We conclude that the telltale craniofacial and dental abnormalities are important for dentists and other clinicians in the diagnosis of osteopetrosis and other genetic bone diseases.
Collapse
Affiliation(s)
- Yu Ma
- College of Life Sciences, Northwest University, Xi'an 710069, China
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Disease, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yali Xu
- College of Life Sciences, Northwest University, Xi'an 710069, China
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Disease, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yanli Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Disease, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiaohong Duan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Disease, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
36
|
Lei Q, Yang J, Li L, Zhao N, Lu C, Lu A, He X. Lipid metabolism and rheumatoid arthritis. Front Immunol 2023; 14:1190607. [PMID: 37325667 PMCID: PMC10264672 DOI: 10.3389/fimmu.2023.1190607] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
As a chronic progressive autoimmune disease, rheumatoid arthritis (RA) is characterized by mainly damaging the synovium of peripheral joints and causing joint destruction and early disability. RA is also associated with a high incidence rate and mortality of cardiovascular disease. Recently, the relationship between lipid metabolism and RA has gradually attracted attention. Plasma lipid changes in RA patients are often detected in clinical tests, the systemic inflammatory status and drug treatment of RA patients can interact with the metabolic level of the body. With the development of lipid metabolomics, the changes of lipid small molecules and potential metabolic pathways have been gradually discovered, which makes the lipid metabolism of RA patients or the systemic changes of lipid metabolism after treatment more and more comprehensive. This article reviews the lipid level of RA patients, as well as the relationship between inflammation, joint destruction, cardiovascular disease, and lipid level. In addition, this review describes the effect of anti-rheumatic drugs or dietary intervention on the lipid profile of RA patients to better understand RA.
Collapse
Affiliation(s)
- Qian Lei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Yang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Onji M, Penninger JM. RANKL and RANK in Cancer Therapy. Physiology (Bethesda) 2023; 38:0. [PMID: 36473204 DOI: 10.1152/physiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Receptor activator of nuclear factor-κB (RANK) and its ligand (RANKL) are key regulators of mammalian physiology such as bone metabolism, immune tolerance and antitumor immunity, and mammary gland biology. Here, we explore the multiple functions of RANKL/RANK in physiology and pathophysiology and discuss underlying principles and strategies to modulate the RANKL/RANK pathway as a therapeutic target in immune-mediated cancer treatment.
Collapse
Affiliation(s)
- Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
38
|
Osteoclastogenesis of human peripheral blood, bone marrow, and cord blood monocytes. Sci Rep 2023; 13:3763. [PMID: 36882450 PMCID: PMC9992388 DOI: 10.1038/s41598-023-30701-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Osteoclasts are multinucleated bone resorbing cells that can be differentiated from human monocytes in vitro. There are few studies comparing osteoclastogenesis of different monocyte sources. We compared monocytes from human bone marrow (BM), peripheral blood (PB), and umbilical cord blood (CB) and their osteoclastogenic potential by culturing them with RANKL (20 and 80 ng/ml) and M-CSF (10 ng/ml) for 14 days. We also cultured cells without growth factors, as umbilical cord blood monocytes have been reported to be able to fuse spontaneously into osteoclasts. The data was analysed on d4, d8, d11, and d14. After culture with RANKL and M-CSF, all types of cell cultures developed TRACP -positive multinuclear cells that were able to form resorption pits on human bone slices. Only occasional multinuclear cells and small infrequent resorbed areas could be found in PB and CB-derived cultures without growth factors. BM-derived cells formed greater resorption areas than PB- and CB-derived monocytes. The greatest monocyte population in BM samples were intermediate (CD14++CD16+) and in PB and CB classical monocytes (76.3% and 54.4%, respectively). In conclusion, our data demonstrates that bone resorbing osteoclasts can be differentiated from BM, PB and CB. However, the osteoclast precursor origin can affect the osteoclast properties and function.
Collapse
|
39
|
Research progress on the role of extracellular vesicles derived from aging cells in osteoporosis. Biosci Rep 2023; 43:232531. [PMID: 36734979 PMCID: PMC9939407 DOI: 10.1042/bsr20221775] [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: 08/15/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/04/2023] Open
Abstract
The occurrence and development of many diseases are highly associated with the aging of the body. Among them, osteoporosis (OP) is a common age-related disease that tends to occur in the elderly population and is highly related to the aging factors in the body. In the process of aging transmission, the senescence-related secretory phenotype (SASP) can convey the information about aging through the paracrine pathway and endocrine mechanism through the extracellular vesicles (EVs) connected to SASP. EVs can be used as a way of conduction to join the connection between micro-environmental aging and age-related illnesses. EVs are double-layer membranous vesicles separated or secreted from the cell membrane, which mainly include microvesicles (MVs) and exosomes. Vesicular bodies secreted by this exocrine form carry a variety of cell-derived related substances (including a variety of proteins, lipids, DNA, mRNA, miRNAs, etc). These substances are mainly concentrated in human body fluids, especially can be transported to all parts of the body with the blood circulation system, and participate in the interactions between cells. Osteoporosis is closely associated with aging and aging cells, suggesting EVs were active in this pathological process. In this article, the basic mechanisms of aging cells in the occurrence and progression of osteoporosis through EVs will be discussed, to explore the connection between aging and osteoporosis, thereby providing a new perspective on the occurrence and development as well as prevention and treatment of osteoporosis.
Collapse
|
40
|
Molecular Basis beyond Interrelated Bone Resorption/Regeneration in Periodontal Diseases: A Concise Review. Int J Mol Sci 2023; 24:ijms24054599. [PMID: 36902030 PMCID: PMC10003253 DOI: 10.3390/ijms24054599] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 03/02/2023] Open
Abstract
Periodontitis is the sixth most common chronic inflammatory disease, destroying the tissues supporting the teeth. There are three distinct stages in periodontitis: infection, inflammation, and tissue destruction, where each stage has its own characteristics and hence its line of treatment. Illuminating the underlying mechanisms of alveolar bone loss is vital in the treatment of periodontitis to allow for subsequent reconstruction of the periodontium. Bone cells, including osteoclasts, osteoblasts, and bone marrow stromal cells, classically were thought to control bone destruction in periodontitis. Lately, osteocytes were found to assist in inflammation-related bone remodeling besides being able to initiate physiological bone remodeling. Furthermore, mesenchymal stem cells (MSCs) either transplanted or homed exhibit highly immunosuppressive properties, such as preventing monocytes/hematopoietic precursor differentiation and downregulating excessive release of inflammatory cytokines. In the early stages of bone regeneration, an acute inflammatory response is critical for the recruitment of MSCs, controlling their migration, and their differentiation. Later during bone remodeling, the interaction and balance between proinflammatory and anti-inflammatory cytokines could regulate MSC properties, resulting in either bone formation or bone resorption. This narrative review elaborates on the important interactions between inflammatory stimuli during periodontal diseases, bone cells, MSCs, and subsequent bone regeneration or bone resorption. Understanding these concepts will open up new possibilities for promoting bone regeneration and hindering bone loss caused by periodontal diseases.
Collapse
|
41
|
Athonvarangkul D, Wysolmerski JJ. Crosstalk within a brain-breast-bone axis regulates mineral and skeletal metabolism during lactation. Front Physiol 2023; 14:1121579. [PMID: 36875035 PMCID: PMC9979219 DOI: 10.3389/fphys.2023.1121579] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
To support the increased calcium demands for milk production during lactation, a dramatic and reversible physiological response occurs to alter bone and mineral metabolism. This coordinated process involves a brain-breast-bone axis that integrates hormonal signals that allow for adequate calcium delivery to milk yet also protects the maternal skeletal from excessive bone loss or decreases in bone quality or function. Here, we review the current knowledge on the crosstalk between the hypothalamus, mammary gland, and skeleton during lactation. We discuss the rare entity of pregnancy and lactation associated osteoporosis and consider how the physiology of bone turnover in lactation may impact the pathophysiology of postmenopausal osteoporosis. Further understanding of the regulators of bone loss during lactation, particularly in humans, may provide insights into new therapies for osteoporosis and other diseases of excess bone loss.
Collapse
Affiliation(s)
- Diana Athonvarangkul
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | | |
Collapse
|
42
|
Zhan W, Ruan B, Dong H, Wang C, Wu S, Yu H, Xu X, Sun H, Cai J. Isopsoralen suppresses receptor activator of nuclear factor kappa- β ligand-induced osteoclastogenesis by inhibiting the NF- κB signaling. PeerJ 2023; 11:e14560. [PMID: 36643647 PMCID: PMC9838210 DOI: 10.7717/peerj.14560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/22/2022] [Indexed: 01/12/2023] Open
Abstract
Osteoporosis is a serious systemic metabolic bone system disease.This study aimed to identify the target genes of isopsoralen and the signaling pathways involved in the differential expression of the genes involved in osteoclast differentiation. We hypothesized that isopsoralen may inhibit osteoclast differentiation by blocking the nuclear factor kappa-B (NF-κB) signaling pathway and verified our hypothesis through basic experiments. The 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was used to detect the effect of isopsoralen on the proliferation and viability of primary mouse bone marrow monocytes (BMMCs). The effect of isopsoralen on receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation was determined by using tartrate-resistant acid phosphatase (TRAP) staining. Quantitative real-time PCR (qRT-PCR) and Western blot were used to detect the expression of the related genes and proteins. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway of isopsoralen target genes were obtained through comprehensive analysis using the STITCH database, Cytoscape 3.8.2, and R-Studio software. Differentially expressed genes (DEGs) were found in osteoclasts induced by RANKL before and after 3 days using R-Studio, following which KEGG analysis was performed. Next, enrichment analysis was performed on the KEGG pathway shared by the target genes of isopsoralen and the differentially expressed genes during osteoclast differentiation to predict the signaling pathway underlying the inhibition of osteoclast differentiation by isopsoralen. Finally, Western blot was used to detect the effect of isopsoralen on the activation of signaling pathways to verify the results of our bioinformatics analysis. Based on the enrichment analysis of isopsoralen target genes and differentially expressed genes during osteoclastogenesis, we believe that isopsoralen can inhibit RANKL-induced osteoclastogenesis by inhibiting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wanda Zhan
- College of Medicine, Yangzhou University, Yangzhou, Jiangsu, China,Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Binjia Ruan
- Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Hui Dong
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chaoyong Wang
- College of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shuangshi Wu
- College of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hang Yu
- College of Medicine, Yangzhou University, Yangzhou, Jiangsu, China,Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaohang Xu
- College of Medicine, Yangzhou University, Yangzhou, Jiangsu, China,Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hao Sun
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun Cai
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
43
|
Wang Y, Zhong Z, Ma M, Zhao Y, Zhang C, Qian Z, Wang B. The role played by ailanthone in inhibiting bone metastasis of breast cancer by regulating tumor-bone microenvironment through the RANKL-dependent pathway. Front Pharmacol 2023; 13:1081978. [PMID: 36686653 PMCID: PMC9849906 DOI: 10.3389/fphar.2022.1081978] [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: 10/27/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction: Bone metastasis of breast cancer (BC) is a process in which the disruption of the bone homeostatic microenvironment leads to an increase in osteoclast differentiation. Ailanthus altissima shows an inhibitory effect on osteoclast differentiation. Ailanthone (AIL) refers to a natural compound isolated from Ailanthus altissima, a Chinese herbal medicine, and has effective anti-tumor activity in numerous cell lines. Its impact on bone metastases for BC is yet unclear. Methods: We measured the effect of AIL on MDA-MB-231 cells by wound healing experiments, Transwell and colony formation experiment. Using the Tartrate-resistant Acid Phosphatase (TRAP) staining tests, filamentous (F-actin) staining and bone resorption test to detect the effect of AIL on the osteoclast cell differentiation of the Bone Marrow-derived Macrophages (BMMs), activated by the MDA-MB-231 cell Conditioned Medium (MDA-MB-231 CM) and the Receptor Activator of Nuclear factor-κB Ligand (RANKL),and to explore its possibility Mechanisms. In vivo experiments verified the effect of AIL on bone destruction in breast cancer bone metastasis model mice. Results: In vitro, AIL significantly decrease the proliferation, migration and infiltration abilities of MDA-MB-231 cells at a safe concentration, and also reduced the expression of genes and proteins involved in osteoclast formation in MDA-MB-231 cells. Osteoclast cell differentiation of the BMMs, activated by MDA-MB-231 CM and RANKL, were suppressed by AIL in the concentration-dependent manner. Additionally, it inhibits osteoclast-specific gene and protein expression. It was noted that AIL inhibited the expression of the osteoclast differentiation-related cytokines RANKL and interleukin-1β (IL-1β) that were secreted by the MDA-MB-231 cells after upregulating the Forkhead box protein 3 (FOXP3) expression. Furthermore, AIL also inhibits the expression of the Mitogen-Activated Protein Kinase (MAPK), Phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), and Nuclear factor-κB Ligand (NF-κB) signaling pathways, which then suppresses the MDA-MB-231CM-induced development of Osteoclasts. Conclusion: Our study shows that AIL blocks osteoclast differentiation in the bone metastasis microenvironment by inhibiting cytokines secreted by BC cells, which may be a potential agent for the treatment of BC and its secondary bone metastasis.
Collapse
Affiliation(s)
- Yajun Wang
- Department of Breast Cancer and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zeyuan Zhong
- Shanghai Medical College, Fudan University, Shanghai, China,Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Miao Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yannan Zhao
- Department of Breast Cancer and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chongjing Zhang
- Shanghai Medical College, Fudan University, Shanghai, China,Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China,*Correspondence: Biyun Wang, ; Zhi Qian, ; Chongjing Zhang,
| | - Zhi Qian
- Institution of Orthopedic Diseases, Zhangye People’s Hospital Affiliated to Hexi University, Zhangye, China,*Correspondence: Biyun Wang, ; Zhi Qian, ; Chongjing Zhang,
| | - Biyun Wang
- Department of Breast Cancer and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,*Correspondence: Biyun Wang, ; Zhi Qian, ; Chongjing Zhang,
| |
Collapse
|
44
|
Bae S, Kim K, Kang K, Kim H, Lee M, Oh B, Kaneko K, Ma S, Choi JH, Kwak H, Lee EY, Park SH, Park-Min KH. RANKL-responsive epigenetic mechanism reprograms macrophages into bone-resorbing osteoclasts. Cell Mol Immunol 2023; 20:94-109. [PMID: 36513810 PMCID: PMC9794822 DOI: 10.1038/s41423-022-00959-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 12/15/2022] Open
Abstract
Monocyte/macrophage lineage cells are highly plastic and can differentiate into various cells under different environmental stimuli. Bone-resorbing osteoclasts are derived from the monocyte/macrophage lineage in response to receptor activator of NF-κB ligand (RANKL). However, the epigenetic signature contributing to the fate commitment of monocyte/macrophage lineage differentiation into human osteoclasts is largely unknown. In this study, we identified RANKL-responsive human osteoclast-specific superenhancers (SEs) and SE-associated enhancer RNAs (SE-eRNAs) by integrating data obtained from ChIP-seq, ATAC-seq, nuclear RNA-seq and PRO-seq analyses. RANKL induced the formation of 200 SEs, which are large clusters of enhancers, while suppressing 148 SEs in macrophages. RANKL-responsive SEs were strongly correlated with genes in the osteoclastogenic program and were selectively increased in human osteoclasts but marginally presented in osteoblasts, CD4+ T cells, and CD34+ cells. In addition to the major transcription factors identified in osteoclasts, we found that BATF binding motifs were highly enriched in RANKL-responsive SEs. The depletion of BATF1/3 inhibited RANKL-induced osteoclast differentiation. Furthermore, we found increased chromatin accessibility in SE regions, where RNA polymerase II was significantly recruited to induce the extragenic transcription of SE-eRNAs, in human osteoclasts. Knocking down SE-eRNAs in the vicinity of the NFATc1 gene diminished the expression of NFATc1, a major regulator of osteoclasts, and osteoclast differentiation. Inhibiting BET proteins suppressed the formation of some RANKL-responsive SEs and NFATc1-associated SEs, and the expression of SE-eRNA:NFATc1. Moreover, SE-eRNA:NFATc1 was highly expressed in the synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential. Our genome-wide analysis revealed RANKL-inducible SEs and SE-eRNAs as osteoclast-specific signatures, which may contribute to the development of osteoclast-specific therapeutic interventions.
Collapse
Affiliation(s)
- Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Kibyeong Kim
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Keunsoo Kang
- Department of Microbiology, Dankook University, Cheonan, 3116, Republic of Korea
| | - Haemin Kim
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Minjoon Lee
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Kaichi Kaneko
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Sungkook Ma
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jae Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, USA
| | - Eun Young Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Sung Ho Park
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10021, USA.
| |
Collapse
|
45
|
The Role of procollagen type 1 amino-terminal propertied (P1NP) Cytochrome P450 (CYPs) and Osteoprotegerin (OPG) as Potential Bone function markers in Prostate Cancer Bone Metastasis. REV ROMANA MED LAB 2023. [DOI: 10.2478/rrlm-2023-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Abstract
Background: Procollagen type I amino-terminal propeptide (PINP) is often present during osteoblast development and could be a biomarker of early bone development. Osteoprotegerin (OPG) may protect tumor cells from apoptosis. Cytochrome P450 enzymes help tumor development and treatment (CYPs). Cytochrome P450 activates and deactivates anticancer drugs and procarcinogens.
Objective: The study examined the amounts of a diagnostic marker of bone formation, the amino terminal propeptide of type I procollagen (PINP), Osteoprotegerin (OPG), and P450, in prostate cancer patients at different stages and its ability to detect osteoblastic metastases.
Methods: ELISA was used to measure PINP, OPG, and P450 levels in 30 prostate cancer patients. (n = 32) and healthy men’s serum (n = 36).
Results: Prostate cancer patients had higher blood levels of PINP, OPG, and P450 than healthy persons (301.3±134.9, 980±467.2, and 84.2±28.4 pg/mL, respectively). Compared to I+II prostate cancer patients, III+IV patients showed higher serum PINP, OPG, and P450 levels (P 0.001). OPG, P450, and PINP had statistically significant Area under the ROC curve (0.9467, P= 0.0001, 0.91, P= 0.0001, and 0.6977, P= 0.4035) in prostate cancer patients.
Conclusions: Metastatic prostate cancer patients had greater PINP, OPG, and P450 levels, according to our findings. PINP, OPG, and P450 levels may affect prostate cancer progression. These findings imply that serum PINP, OPG, and P450 levels may predict and diagnose prostate cancer.
Collapse
|
46
|
Sun J, Cai G, Shen J, Cheng P, Zhang J, Jiang D, Xu X, Lu F, Chen L, Chen H. AS-605240 Blunts Osteoporosis by Inhibition of Bone Resorption. Drug Des Devel Ther 2023; 17:1275-1288. [PMID: 37138583 PMCID: PMC10150757 DOI: 10.2147/dddt.s403231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/19/2023] [Indexed: 05/05/2023] Open
Abstract
Background Osteoporosis is a metabolic bone disease. Osteoclasts are significantly involved in the pathogenesis of osteoporosis. AS-605240 (AS) is a small molecule PI3K-γ inhibitor and is less toxic compared to pan-PI3K inhibitors. AS also exerts multiple biological effects including anti-inflammatory, anti-tumor, and myocardial remodeling promotion. However, the involvement of AS in the differentiation and functions of osteoclasts and the effect of AS in treating patients with osteoporosis is still unclear. Purpose This study aimed to investigate if AS inhibits the differentiation of osteoclasts and resorption of the bones induced by M-CSF and RANKL. Next, we evaluated the therapeutic effects of AS on bone loss in ovariectomy (OVX)-induced osteoporosis mice models. Methods We stimulated bone marrow-derived macrophages with an osteoclast differentiation medium containing different AS concentrations for 6 days or 5μM AS at different times. Next, we performed tartrate-resistant acid phosphatase (TRAP) staining, bone resorption assay, F-actin ring fluorescence, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting (WB). Next, MC3T3-E1s (pre-osteoblast cells) were differentiated to osteoblast by stimulating the cells with varying AS concentrations. Next, we performed alkaline phosphatase (ALP) staining, RT-qPCR, and WB on these cells. We established an OVX-induced osteoporosis mice model and treated the mice with 20mg/kg of AS. Finally, we extracted the femurs and performed micro-CT scanning, H&E, and TRAP staining. Results AS inhibits the formation of osteoclasts and resorption of bone triggered by RANKL by inhibiting the PI3K/Akt signaling pathway. Furthermore, AS enhances the differentiation of osteoblasts and inhibits bone loss due to OVX in vivo. Conclusion AS inhibits osteoclast production and enhances osteoblast differentiation in mice, thus providing a new therapeutic approach for treating patients with osteoporosis.
Collapse
Affiliation(s)
- Jiacheng Sun
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Guoping Cai
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Jinlong Shen
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Pu Cheng
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Jiapeng Zhang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Dengteng Jiang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Xianquan Xu
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Fangying Lu
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Lihua Chen
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
| | - Haixiao Chen
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, People’s Republic of China
- Bone Development and Metabolism Research Center of Taizhou Hospital, Linhai, Zhejiang Province, People’s Republic of China
- Correspondence: Haixiao Chen; Lihua Chen, Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, N.150 Ximen Road of Linhai City, Taizhou, Zhejiang Province, People’s Republic of China, Tel +86 15268400288, +86 13757624851, Email ;
| |
Collapse
|
47
|
Yi X, Hu G, Yang Y, Li J, Jin J, Chang B. Role of MOTS-c in the regulation of bone metabolism. Front Physiol 2023; 14:1149120. [PMID: 37200834 PMCID: PMC10185875 DOI: 10.3389/fphys.2023.1149120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/18/2023] [Indexed: 05/20/2023] Open
Abstract
MOTS-c, a mitochondrial-derived peptide (MDP), is an essential regulatory mediator of cell protection and energy metabolism and is involved in the development of specific diseases. Recent studies have revealed that MOTS-c promotes osteoblast proliferation, differentiation, and mineralization. Furthermore, it inhibits osteoclast production and mediates the regulation of bone metabolism and bone remodeling. Exercise effectively upregulates the expression of MOTS-c, but the specific mechanism of MOTS-c regulation in bone by exercise remains unclear. Therefore, this article reviewed the distribution and function of MOTS-c in the tissue, discussed the latest research developments in the regulation of osteoblasts and osteoclasts, and proposed potential molecular mechanisms for the effect of exercise on the regulation of bone metabolism. This review provides a theoretical reference for establishing methods to prevent and treat skeletal metabolic diseases.
Collapse
Affiliation(s)
- Xuejie Yi
- Social Science Research Center, Shenyang Sport University, Shenyang, Liaoning, China
| | - Guangxuan Hu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yang Yang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jing Li
- School of Physical Education, Liaoning Normal University, Dalian, Liaoning, China
| | - Junjie Jin
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Bo Chang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
- *Correspondence: Bo Chang,
| |
Collapse
|
48
|
Zhang Y, Liang J, Liu P, Wang Q, Liu L, Zhao H. The RANK/RANKL/OPG system and tumor bone metastasis: Potential mechanisms and therapeutic strategies. Front Endocrinol (Lausanne) 2022; 13:1063815. [PMID: 36589815 PMCID: PMC9800780 DOI: 10.3389/fendo.2022.1063815] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
With the markedly increased diagnosis and incidence of cancer in the population, tumor bone metastasis has become a frequent event in tumor patients. Healthy bone integrity is maintained by a delicate balance between bone formation and bone resorption. Unfortunately, many tumors, such as prostate and breast, often metastasize to the bone, and the alterations to the bone homeostasis can particularly favor tumor homing and consequent osteolytic or osteoblastic lesions. Receptor activator of NF-κB ligand (RANKL), its receptor RANK, and osteoprotegerin (OPG) are involved in the regulation of the activation, differentiation, and survival of osteoclasts, which play critical roles in bone metastasis formation. High rates of osteoclastic bone resorption significantly increase fracture risk, cause severe bone pain, and contribute to homing tumor cells in bone and bone marrow. Consequently, suppression of the RANK/RANKL/OPG system and osteoclastic activity can not only ameliorate bone resorption but may also prevent tumor bone metastases. This review summarizes the important role of the RANK/RANKL/OPG system and osteoclasts in bone homeostasis and its effect on tumor bone metastasis and discusses therapeutic strategies based on RANKL inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
49
|
Qu Z, Zhang B, Kong L, Gong Y, Feng M, Gao X, Wang D, Yan L. Receptor activator of nuclear factor-κB ligand-mediated osteoclastogenesis signaling pathway and related therapeutic natural compounds. Front Pharmacol 2022; 13:1043975. [PMID: 36438811 PMCID: PMC9683337 DOI: 10.3389/fphar.2022.1043975] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Osteoclast is a hematopoietic precursor cell derived from the mononuclear macrophage cell line, which is the only cell with bone resorption function. Its abnormal activation can cause serious osteolysis related diseases such as rheumatoid arthritis, Paget's disease and osteoporosis. In recent years, the adverse effects caused by anabolic anti-osteolytic drugs have increased the interest of researchers in the potential therapeutic and preventive effects of natural plant derivatives and natural compounds against osteolytic diseases caused by osteoclasts. Natural plant derivatives and natural compounds have become major research hotspots for the treatment of osteolysis-related diseases due to their good safety profile and ability to improve bone. This paper provides an overview of recent advances in the molecular mechanisms of RANKL and downstream signaling pathways in osteoclast differentiation, and briefly outlines potential natural compounds with antiosteoclast activity and molecular mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
50
|
Abstract
Osteopetrosis (OPT) is a rare inherited bone disease characterized by a bone resorption defect, due to osteoclast malfunction (in osteoclast-rich, oc-rich, OPT forms) or absence (in oc-poor OPT forms). This causes severe clinical abnormalities, including increased bone density, lack of bone marrow cavity, stunted growth, macrocephaly, progressive deafness, blindness, hepatosplenomegaly, and severe anemia. The oc-poor subtype of OPT is ultra-rare in humans. It is caused by mutations in either the tumor necrosis factor ligand superfamily member 11 (TNFSF11) gene, encoding RANKL (Receptor Activator of Nuclear factor-kappa B [NF-κB] Ligand) which is expressed on cells of mesenchymal origin and lymphocytes, or the TNFRSF member 11A (TNFRSF11A) gene, encoding the RANKL functional receptor RANK which is expressed on cells of myeloid lineage including osteoclasts. Clinical presentation is usually severe with onset in early infancy or in fetal life, although as more patients are reported, expressivity is variable. Phenotypic variability of RANK-deficient OPT sometimes includes hypogammaglobulinemia or radiological features of dysosteosclerosis. Disease progression is somewhat slower in RANKL-deficient OPT than in other 'malignant' subtypes of OPT. While both RANKL and RANK are essential for normal bone turnover, hematopoietic stem cell transplantation (HSCT) is the treatment of choice only for patients with the RANK-deficient form of oc-poor OPT. So far, there is no cure for RANKL-deficient OPT.
Collapse
Affiliation(s)
- Cristina Sobacchi
- CNR-IRGB, Milan Unit, via Fantoli 16/15, 20138 Milan, Italy; Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, MI, Italy.
| | - Mario Abinun
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|