1
|
Qin Y, Shirakawa J, Xu C, Chen R, Yang X, Ng C, Nakano S, Elguindy M, Deng Z, Prasanth KV, Eissmann MF, Nakagawa S, Ricci WM, Zhao B. Long non-coding RNA Malat1 fine-tunes bone homeostasis and repair by orchestrating cellular crosstalk and β-catenin-OPG/Jagged1 pathway. eLife 2024; 13:RP98900. [PMID: 39714456 DOI: 10.7554/elife.98900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts, and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.
Collapse
Affiliation(s)
- Yongli Qin
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Jumpei Shirakawa
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Ruge Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, United States
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, United States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Shinichi Nakano
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Mahmoud Elguindy
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer center at Illinois, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Moritz F Eissmann
- Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - William M Ricci
- Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, NewYork, United States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, United States
| |
Collapse
|
2
|
Venkataraman A, Kordic I, Li J, Zhang N, Bharadwaj NS, Fang Z, Das S, Coskun AF. Decoding senescence of aging single cells at the nexus of biomaterials, microfluidics, and spatial omics. NPJ AGING 2024; 10:57. [PMID: 39592596 PMCID: PMC11599402 DOI: 10.1038/s41514-024-00178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Aging has profound effects on the body, most notably an increase in the prevalence of several diseases. An important aging hallmark is the presence of senescent cells that no longer multiply nor die off properly. Another characteristic is an altered immune system that fails to properly self-surveil. In this multi-player aging process, cellular senescence induces a change in the secretory phenotype, known as senescence-associated secretory phenotype (SASP), of many cells with the intention of recruiting immune cells to accelerate the clearance of these damaged senescent cells. However, the SASP phenotype results in inducing secondary senescence of nearby cells, resulting in those cells becoming senescent, and improper immune activation resulting in a state of chronic inflammation, called inflammaging, in many diseases. Senescence in immune cells, termed immunosenescence, results in further dysregulation of the immune system. An interdisciplinary approach is needed to physiologically assess aging changes of the immune system at the cellular and tissue level. Thus, the intersection of biomaterials, microfluidics, and spatial omics has great potential to collectively model aging and immunosenescence. Each of these approaches mimics unique aspects of the body undergoes as a part of aging. This perspective highlights the key aspects of how biomaterials provide non-cellular cues to cell aging, microfluidics recapitulate flow-induced and multi-cellular dynamics, and spatial omics analyses dissect the coordination of several biomarkers of senescence as a function of cell interactions in distinct tissue environments. An overview of how senescence and immune dysregulation play a role in organ aging, cancer, wound healing, Alzheimer's, and osteoporosis is included. To illuminate the societal impact of aging, an increasing trend in anti-senescence and anti-aging interventions, including pharmacological interventions, medical procedures, and lifestyle changes is discussed, including further context of senescence.
Collapse
Affiliation(s)
- Abhijeet Venkataraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ivan Kordic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - JiaXun Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nivik Sanjay Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandip Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Garcez-Filho J, Bianco DDA, Barbisan de Souza A, de Oliveira RP, Matarazzo F, Araújo MG. Alveolar ridge bone changes in patients treated with the split-crest technique with simultaneous implant placement: A retrospective longitudinal observational study. Clin Oral Implants Res 2024; 35:1440-1451. [PMID: 39076119 DOI: 10.1111/clr.14331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 06/06/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVES To investigate long-term alveolar ridge bone changes in patients treated with the split-crest technique (SCT) with simultaneous implant placement. MATERIALS AND METHODS Alveolar ridge width (ARW) was measured with a caliper immediately before (ARW1) and after SCT (ARW2) with a caliper. Existing CBCT scans taken at least 5 years postoperatively were used to assess the healed ARW (ARWF), buccal bone thickness (BBT), and lingual/palatal bone thickness (L/PBT) at 0, 3, and 5 mm apically from implant platform level (IPL). The distance between IPL and buccal bone crest (IPL-BBC) was also measured. Findings were compared with Wilcoxon and Student's t tests (p < .05). RESULTS Records of thirty patients with 85 implants were included. Eleven patients (31 implants) presented CBCT scans taken in two separate occasions, resulting in 116 measurements. Tomographic follow-up ranged from 5 to 18 years (11.5 ± 4.18 years). Mean ARW increased from 3.2 ± 0.6 to 6.6 ± 0.48 mm after SCT, but significantly reduced overtime to 4.46 ± 0.83 mm (ARWF) (p < .0001). Mean BBT was 0.5 ± 0.9, 1.3 ± 0.9, and 2.0 ± 1.2 mm, while mean L/PBT was 0.9 ± 0.7, 1.6 ± 0.9, and 2.1 ± 1.0 mm at 0, 3, and 5 mm from IPL, respectively. IPL-BBC distance varied up to 8.7 mm (1.4 ± 2.0 mm), with 56/116 measurements (51/85 implants) showing IPL-BBC = 0 mm. CONCLUSION Although SCT with simultaneous implant placement can predictably increase ARW postoperatively, ARW tended to significantly reduce over time, due to reductions in BBT and L/PBT, especially coronally. Nonetheless, 60% of the implants still presented some buccal bone at IPL.
Collapse
Affiliation(s)
| | | | - André Barbisan de Souza
- Department of Dentistry, Tufts University School of Dental Medicine, Boca Raton, Florida, USA
| | | | - Flávia Matarazzo
- Department of Dentistry, State University of Maringá, Maringá, Brazil
| | - Maurício G Araújo
- Department of Dentistry, State University of Maringá, Maringá, Brazil
| |
Collapse
|
4
|
Qin Y, Shirakawa J, Xu C, Chen R, Yang X, Ng C, Nakano S, Elguindy M, Deng Z, Prasanth KV, Eissmann MF, Nakagawa S, Ricci WM, Zhao B. Long non-coding RNA Malat1 fine-tunes bone homeostasis and repair by orchestrating cellular crosstalk and the β-catenin-OPG/Jagged1 pathway. RESEARCH SQUARE 2024:rs.3.rs-3793919. [PMID: 38234849 PMCID: PMC10793491 DOI: 10.21203/rs.3.rs-3793919/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.
Collapse
Affiliation(s)
- Yongli Qin
- Arthritis and Tissue Degeneration Program and 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
| | - Jumpei Shirakawa
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Ruge Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Shinichi Nakano
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Mahmoud Elguindy
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Moritz F. Eissmann
- Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, 60596 Frankfurt, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - William M. Ricci
- Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and 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
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| |
Collapse
|
5
|
Kim HJ, Choi SA, Gu MS, Ko SY, Kwon JH, Han JY, Kim JH, Kim MG. Effects of Glucagon-Like Peptide-1 Receptor Agonist on Bone Mineral Density and Bone Turnover Markers: A Meta-Analysis. Diabetes Metab Res Rev 2024; 40:e3843. [PMID: 39311048 DOI: 10.1002/dmrr.3843] [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: 06/13/2023] [Revised: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024]
Abstract
AIMS Glucagon-like peptide-1 receptor agonist (GLP-1RA) may promote bone formation, but conversely, they could also weaken bones due to the reduction in mechanical load associated with weight loss. However, the clinical effects in humans have not been clearly demonstrated. This meta-analysis aimed to evaluate whether GLP-1RAs affect BMD and bone turnover markers. MATERIAL AND METHODS PubMed, Embase, and Scopus were searched on June 13, 2024. The eligibility criteria were: (1) human studies, (2) receiving a GLP-1RA for more than 4 weeks, (3) an untreated control group or a placebo group, (4) reporting of at least one BMD or bone turnover marker, and (5) an RCT design. The risk of bias was assessed using the Cochrane risk of bias 2 tool. Fixed- or random-effects meta-analysis was performed according to heterogeneity. RESULTS Seven studies were included in the meta-analysis. GLP-1RAs did not significantly change BMD in the femoral neck (mean difference [MD], 0.01 g/cm2; 95% CI, -0.01-0.04 g/cm2), in the total hip (MD, -0.01 g/cm2; 95% CI, -0.02-0.01 g/cm2), and in the lumbar spine (MD, 0 g/cm2; 95% CI, -0.02-0.02 g/cm2). C-terminal telopeptide of type 1 collagen (CTX), a bone resorption marker, significantly increased after GLP-1RA treatment (MD, 0.04 μg/L; 95% CI, 0.01-0.07 μg/L). GLP-1RAs did not significantly change bone formation markers such as procollagen type 1 N-terminal propeptide, bone-specific alkaline phosphatase, osteocalcin. CONCLUSIONS GLP-1RA did not affect BMD and bone formation markers. However, GLP-1RAs led to a significant increase in CTX.
Collapse
Affiliation(s)
- Hee-Ju Kim
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seo-A Choi
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Min-Sun Gu
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seo-Yeong Ko
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Jae-Hee Kwon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Ja-Young Han
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, Republic of Korea
| | - Myeong Gyu Kim
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Stephan CN, Caple JM, D'Alonzo Jaques SS, Byrd JE. Cervicothoracic junction in disaster victim identification: Idiosyncrasies and relevance of body position for advanced chest radiograph comparisons. J Forensic Sci 2024; 69:1587-1603. [PMID: 38037703 DOI: 10.1111/1556-4029.15436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Standard plain film medical radiographs often form a valuable line of evidence to identify individuals in large-scale fatality events. While commonly available, chest radiographs present a challenge that their analysis is somewhat more involved and complex than radiographic records of other body regions. For example, chest radiographs concern subtler morphological varieties of smaller anatomical features across a larger number of skeletal elements in contrast to frontal sinus comparisons that concern a large, (often) single, highly variable void within one bone. This does not detract from or discount chest radiographs as useful identification aids, but it does demand additional prerequisite skills in radiographic interpretation to ensure valid conclusions are attained. When subjects deviate from standardized antemortem (AM) radiographic positions and/or the image quality decreases, the complexity of a chest radiograph comparison is elevated. Generally, the current body of forensic radiographic comparison literature infrequently addresses these more complex circumstances. In this paper, we use real-world radiographic comparison reference images from a military DVI repatriation context to illustrate these factors and outline some procedures that enable these complexities to be easily recognized and appropriately addressed at case examination. A report for an exemplar case that concurrently highlights multiple factors is presented. For novices learning radiographic comparison methods, this case review saliently demonstrates: (1) why the AM reference radiograph(s) drive(s) the radiographic comparison procedure; (2) why care should be taken for correct positioning of the cervicothoracic junction in postmortem radiography of chest elements.
Collapse
Affiliation(s)
- Carl N Stephan
- Laboratory for Human Craniofacial and Skeletal Identification (HuCS-ID Lab), School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jodi M Caple
- SNA International Supporting the Defense POW/MIA Accounting Agency, Hickam, Hawaii, USA
| | | | - John E Byrd
- Defense POW/MIA Accounting Agency, Hickam, Hawaii, USA
| |
Collapse
|
7
|
Wang F, Li H, Yi K, Wu Y, Bian Q, Guo B, Luo X, Kang Y, Wu Q, Ma Q. Long-term second-generation antipsychotics decreases bone formation and resorption in male patients with schizophrenia. Psychopharmacology (Berl) 2024; 241:1771-1780. [PMID: 38647696 DOI: 10.1007/s00213-024-06592-y] [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: 11/09/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
RATIONALE Patients with schizophrenia with second-generation antipsychotics (SGAs) treatment have shown an increased risk of bone fragility and susceptibility to fracture; however, it is still unclear whether this risk is derived from the effect of antipsychotics on balance of bone metabolism. OBJECTIVES We investigated the changes of two bone turnover biomarkers (BTMs) concentrations in people with schizophrenia receiving SGAs: procollagen type I aminoterminal propeptide (PINP) and C-terminal telopeptide of type I collagen (CTX-1) as BTMs of osteogenesis and bone resorption, respectively, to explore how antipsychotics contribute to bone fragility. METHODS We recruited 59 Chinese male patients with schizophrenia (32 drug-naïve first-episode (DNFE) patients and 27 chronic patients) to undergo 8 weeks SGAs treatment. Fasting peripheral blood samples of pre- and posttreatment were collected, plasma levels of PINP and CTX-1 were measured. RESULTS The interaction effects of group and time on PINP and CTX-1 concentrations were found (P = .016 and P = .008). There was a significant decrease for both BTMs concentrations of the posttreatment compared to the pretreatment (P<.001 and P = .003). Chronic patients had significantly higher changes of BTMs concentrations compared to DNFE patients (P = .048 and P = .024). There was a positive correlation of the two BTMs of pretreatment with disease course in DNFE group (r = .37, P = .039;r = .38, P = .035) and a negative correlation of PINP of pretreatment with age in the chronic group (r=-.40, P = .039). CONCLUSION Long-term SGAs medication inhibited osteogenesis in a dose- and time-dependent manner and damaged the balance of bone formation and bone resorption.
Collapse
Affiliation(s)
- Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China.
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China.
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China.
| | - Hui Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Kaijun Yi
- Department of Orthopedics, Xiangyang No. 1 People's Hospital Affiliated to Hubei University of Medicine, Xiangyang, 441000, Hubei, China
| | - Yan Wu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Qingtao Bian
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Baoyan Guo
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Qi Wu
- Fenyang College, Shanxi Medical University, Lvliang, 032200, China
- Department of Psychiatry, Changzhou Peace Hospital, The 102nd Hospital of The Chinese People's Liberation Army, Changzhou, 213003, China
| | - Qinghe Ma
- Department of Psychiatry, Changzhou Peace Hospital, The 102nd Hospital of The Chinese People's Liberation Army, Changzhou, 213003, China
- Department of Internal Medicine, The 904th Hospital of The Chinese People's Liberation Army, Wuxi, 214004, China
| |
Collapse
|
8
|
Grass DM, Malek G, Taïeb HM, Ittah E, Richard H, Reznikov N, Laverty S. Characterization and quantification of in-vitro equine bone resorption in 3D using μCT and deep learning-aided feature segmentation. Bone 2024; 185:117131. [PMID: 38777311 DOI: 10.1016/j.bone.2024.117131] [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: 02/08/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
High cyclic strains induce formation of microcracks in bone, triggering targeted bone remodeling, which entails osteoclastic resorption. Racehorse bone is an ideal model for studying the effects of high-intensity loading, as it is subject to focal formation of microcracks and subsequent bone resorption. The volume of resorption in vitro is considered a direct indicator of osteoclast activity but indirect 2D measurements are used more often. Our objective was to develop an accurate, high-throughput method to quantify equine osteoclast resorption volume in μCT 3D images. Here, equine osteoclasts were cultured on equine bone slices and imaged with μCT pre- and postculture. Individual resorption events were then isolated and analyzed in 3D. Modal volume, maximum depth, and aspect ratio of resorption events were calculated. A convolutional neural network (CNN U-Net-like) was subsequently trained to identify resorption events on post-culture μCT images alone, without the need for pre-culture imaging, using archival bone slices with known resorption areas and paired CTX-I biomarker levels in culture media. 3D resorption volume measurements strongly correlated with both the CTX-I levels (p < 0.001) and area measurements (p < 0.001). Our 3D analysis shows that the shapes of resorption events form a continuous spectrum, rather than previously reported pit and trench categories. With more extensive resorption, shapes of increasing complexity appear, although simpler resorption cavity morphologies (small, rounded) remain most common, in acord with the left-hand limit paradigm. Finally, we show that 2D measurements of in vitro osteoclastic resorption are a robust and reliable proxy.
Collapse
Affiliation(s)
- Debora M Grass
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Gwladys Malek
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Hubert M Taïeb
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Eran Ittah
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Hélène Richard
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Natalie Reznikov
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Sheila Laverty
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
9
|
Soloviova M, Beltrán-Vargas JC, Castro LFD, Belmonte-Beitia J, Pérez-García VM, Caballero M. A Mathematical Model for Fibrous Dysplasia: The Role of the Flow of Mutant Cells. Bull Math Biol 2024; 86:108. [PMID: 39007985 DOI: 10.1007/s11538-024-01336-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024]
Abstract
Fibrous dysplasia (FD) is a mosaic non-inheritable genetic disorder of the skeleton in which normal bone is replaced by structurally unsound fibro-osseous tissue. There is no curative treatment for FD, partly because its pathophysiology is not yet fully known. We present a simple mathematical model of the disease incorporating its basic known biology, to gain insight on the dynamics of the involved bone-cell populations, and shed light on its pathophysiology. We develop an analytical study of the model and study its basic properties. The existence and stability of steady states are studied, an analysis of sensitivity on the model parameters is done, and different numerical simulations provide findings in agreement with the analytical results. We discuss the model dynamics match with known facts on the disease, and how some open questions could be addressed using the model.
Collapse
Affiliation(s)
- Mariia Soloviova
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Avda. Camilo José Cela 3, Ciudad Real, 13071, Spain.
| | - Juan C Beltrán-Vargas
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Avda. Camilo José Cela 3, Ciudad Real, 13071, Spain
| | - Luis Fernandez de Castro
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, USA
| | - Juan Belmonte-Beitia
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Avda. Camilo José Cela 3, Ciudad Real, 13071, Spain
| | - Víctor M Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Avda. Camilo José Cela 3, Ciudad Real, 13071, Spain
| | - Magdalena Caballero
- Department of Mathematics, Universidad de Córdoba, Campus de Rabanales, Córdoba, 14071, Spain
| |
Collapse
|
10
|
Shen Z, Zhang X, Wang Y, Zhu R, Ge L, Cai G. Factors associated with trajectories of bone marrow lesions over 4 years: data from the Osteoarthritis Initiative. Skeletal Radiol 2024; 53:1333-1341. [PMID: 38244061 PMCID: PMC11093866 DOI: 10.1007/s00256-024-04579-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/19/2023] [Accepted: 01/07/2024] [Indexed: 01/22/2024]
Abstract
OBJECTIVE To identify bone marrow lesion (BML) trajectories over 4 years and their demographic and structural predictors in middle-aged and older adults with or at increased risk of knee osteoarthritis (OA). METHODS A total of 614 participants (mean age 61 years, 62% female) from the Osteoarthritis Initiative cohort (OAI) were included. BMLs in 15 anatomical locations of the knee were measured annually from baseline to 4 years using the Magnetic Resonance Imaging Osteoarthritis Knee Score (MOAKS) method. BML trajectories were determined using latent class mixed models (LCMMs). Multinomial logistic regression was used to examine baseline characteristics that predicted BML trajectories. RESULTS Three distinct BML trajectories were identified: "Mild-stable BMLs" (25.9%), "Moderate-stable BMLs" (66.4%), and "Rapid-rise BMLs" (7.7%). Compared to the "Mild-stable BMLs" trajectory, current smokers were more likely to be in the "Moderate-stable BMLs" (odds ratio [OR] 2.089, P < 0.001) and "Rapid-rise" (OR 2.462, P < 0.001) trajectories. Moreover, female sex and meniscal tears were associated with an increased risk of being in the "Rapid-rise BMLs" trajectory (OR 2.023 to 2.504, P < 0.05). Participants who had higher education levels and drank more alcohol were more likely to be in the "Rapid-rise BMLs" trajectory (OR 1.624 to 3.178, P < 0.05) and less likely to be in the "Moderate-stable BMLs" trajectory (OR 0.668 to 0.674, P < 0.05). CONCLUSIONS During the 4-year follow-up, most participants had relatively stable BMLs, few had enlarged BMLs, and no trajectory of decreased BMLs was identified. Sociodemographic factors, lifestyle, and knee structural pathology play roles in predicting distinct BML trajectories.
Collapse
Affiliation(s)
- Ziyuan Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiaoyue Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yining Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Rui Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Liru Ge
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guoqi Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia.
| |
Collapse
|
11
|
Kang T, Yang Z, Zhou M, Lan Y, Hong Y, Gong X, Wu Y, Li M, Chen X, Zhang W. The role of the Piezo1 channel in osteoblasts under cyclic stretching: A study on osteogenic and osteoclast factors. Arch Oral Biol 2024; 163:105963. [PMID: 38608563 DOI: 10.1016/j.archoralbio.2024.105963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/10/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES Orthodontic tooth movement is a mechanobiological reaction induced by appropriate forces, including bone remodeling. The mechanosensitive Piezo channels have been shown to contribute to bone remodeling. However, information about the pathways through which Piezo channels affects osteoblasts remains limited. Thus, we aimed to investigate the influence of Piezo1 on the osteogenic and osteoclast factors in osteoblasts under mechanical load. MATERIALS AND METHODS Cyclic stretch (CS) experiments on MC3T3-E1 were conducted using a BioDynamic mechanical stretching device. The Piezo1 channel blocker GsMTx4 and the Piezo1 channel agonist Yoda1 were used 12 h before the application of CS. MC3T3-E1 cells were then subjected to 15% CS, and the expression of Piezo1, Piezo2, BMP-2, OCN, Runx2, RANKL, p-p65/p65, and ALP was measured using quantitative real-time polymerase chain reaction, western blot, alkaline phosphatase staining, and immunofluorescence staining. RESULTS CS of 15% induced the highest expression of Piezo channel and osteoblast factors. Yoda1 significantly increased the CS-upregulated expression of Piezo1 and ALP activity but not Piezo2 and RANKL. GsMTx4 downregulated the CS-upregulated expression of Piezo1, Piezo2, Runx2, OCN, p-65/65, and ALP activity but could not completely reduce CS-upregulated BMP-2. CONCLUSIONS The appropriate force is more suitable for promoting osteogenic differentiation in MC3T3-E1. The Piezo1 channel participates in osteogenic differentiation of osteoblasts through its influence on the expression of osteogenic factors like BMP-2, Runx2, and OCN and is involved in regulating osteoclasts by influencing phosphorylated p65. These results provide a foundation for further exploration of osteoblast function in orthodontic tooth movement.
Collapse
Affiliation(s)
- Ting Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Ziyuan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Mengqi Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yanhua Lan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yaya Hong
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xinyi Gong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yongjia Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Min Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Weifang Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
12
|
Cheng X, Tian W, Yang J, Wang J, Zhang Y. Engineering approaches to manipulate osteoclast behavior for bone regeneration. Mater Today Bio 2024; 26:101043. [PMID: 38600918 PMCID: PMC11004223 DOI: 10.1016/j.mtbio.2024.101043] [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: 01/03/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Extensive research has delved into the multifaceted roles of osteoclasts beyond their traditional function in bone resorption in recent years, uncovering their significant influence on bone formation. This shift in understanding has spurred investigations into engineering strategies aimed at leveraging osteoclasts to not only inhibit bone resorption but also facilitate bone regeneration. This review seeks to comprehensively examine the mechanisms by which osteoclasts impact bone metabolism. Additionally, it explores various engineering methodologies, including the modification of bioactive material properties, localized drug delivery, and the introduction of exogenous cells, assessing their potential and mechanisms in aiding bone repair by targeting osteoclasts. Finally, the review proposes current limitations and future routes for manipulating osteoclasts through biological and material cues to facilitate bone repair.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, 1098 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
| | - Wenzhi Tian
- Jilin University, Jilin Province Key Lab Tooth Dev & Bone Remodeling, School and Hospital of Stomatology, Department of Oral Pathology, Changchun 130041, Jilin Province, China
| | - Jianhua Yang
- Longgang District People's Hospital of Shenzhen & the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong province, China
| | - Jiamian Wang
- National Innovation Center for Advanced Medical Devices, Shenzhen 518000, Guangdong Province, China
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, 1088 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
- School of Biomedical Engineering, Shenzhen University Medical School, 1088 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
| |
Collapse
|
13
|
Rong S, Li H, Wei Y, Feng Z, Gan L, Deng Z, Zhao L. [Zinc finger protein-36 deficiency inhibits osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells and preosteoblasts by activating the ERK/MAPK pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:697-705. [PMID: 38708503 DOI: 10.12122/j.issn.1673-4254.2024.04.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE To explore the role of zinc finger protein 36(ZFP36) in regulating osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) and preosteoblasts. METHODS ZFP36 expression was observed in primary mouse BMSCs and mouse preosteoblasts (MC3T3-E1 cells) during induced osteogenic differentiation. Zfp36-deficient cell models were constructed in the two cells using RNA interference technique and the changes in differentiation capacities of the transfected cells into osteoblasts were observed. Transcriptome sequencing was used to investigate the potential mechanisms of ZFP36 for regulating osteoblast differentiation of the two cells. U0126, a ERK/MAPK signal suppressor, was used to verify the regulatory mechanism of Zfp36 in osteogenic differentiation of Zfp36-deficient cells. RESULTS During the 14-day induction of osteogenic differentiation, both mouse BMSCs and MC3T3-E1 cells exhibited increased expression of ZFP36, and its mRNA expression reached the peak level on Day 7(P < 0.0001). The Zfp36-deficient cell models showed reduced intensity of alkaline phosphatase (ALP) staining and alizarin red staining with significantly lowered expressions of the osteogenic marker genes including Alpl, Sp7, Bglap and Ibsp (P < 0.01). Transcriptome sequencing verified the reduction of bone mineralization-related gene expressions in Zfp36-deficient cells and indicated the involvement of ERK signaling in the potential regulatory mechanism of Zfp36. Immunoblotting showed that pERK protein expression increased significantly in Zfp36-deficient cells compared with the control cells. In Zfp36-deficient MC3T3-E1 cells, inhibition of activated ERK/MAPK signaling with U0126 resulted in obviously enhanced ALP staining and significantly increased expressions of osteoblast differentiation markers Runx2 and Bglap (P < 0.05). CONCLUSIONS ZFP36 is involved in the regulation of osteoblast differentiation of mouse BMSCs and preosteoblasts, and ZFP36 deficiency causes inhibition of osteoblast differentiation of the cells by activating the ERK/MAPK signaling pathway.
Collapse
Affiliation(s)
- S Rong
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Li
- Beijing Yijiandian Clinic, Beijing 100033, China
- Health Management Center, Peking University International Hospital, Beijing 102206, China
| | - Y Wei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Z Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - L Gan
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Z Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - L Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
14
|
Daponte V, Henke K, Drissi H. Current perspectives on the multiple roles of osteoclasts: Mechanisms of osteoclast-osteoblast communication and potential clinical implications. eLife 2024; 13:e95083. [PMID: 38591777 PMCID: PMC11003748 DOI: 10.7554/elife.95083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
Bone remodeling is a complex process involving the coordinated actions of osteoblasts and osteoclasts to maintain bone homeostasis. While the influence of osteoblasts on osteoclast differentiation is well established, the reciprocal regulation of osteoblasts by osteoclasts has long remained enigmatic. In the past few years, a fascinating new role for osteoclasts has been unveiled in promoting bone formation and facilitating osteoblast migration to the remodeling sites through a number of different mechanisms, including the release of factors from the bone matrix following bone resorption and direct cell-cell interactions. Additionally, considerable evidence has shown that osteoclasts can secrete coupling factors known as clastokines, emphasizing the crucial role of these cells in maintaining bone homeostasis. Due to their osteoprotective function, clastokines hold great promise as potential therapeutic targets for bone diseases. However, despite long-standing work to uncover new clastokines and their effect in vivo, more substantial efforts are still required to decipher the mechanisms and pathways behind their activity in order to translate them into therapies. This comprehensive review provides insights into our evolving understanding of the osteoclast function, highlights the significance of clastokines in bone remodeling, and explores their potential as treatments for bone diseases suggesting future directions for the field.
Collapse
Affiliation(s)
- Valentina Daponte
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| | - Katrin Henke
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| |
Collapse
|
15
|
Long K, Gong A, Yu D, Dong S, Ying Z, Zhang L. Exploring the immunological landscape of osteomyelitis through mendelian randomization analysis. Front Genet 2024; 15:1362432. [PMID: 38650858 PMCID: PMC11033344 DOI: 10.3389/fgene.2024.1362432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
Background Osteomyelitis is a severe bone marrow infection, whose pathogenesis is not yet fully understood. This study aims to explore the causal relationship between immune cell characteristics and osteomyelitis, hoping to provide new insights for the prevention and treatment of osteomyelitis. Methods Based on two independent samples, this study employed a two-sample Mendelian randomization (MR) analysis to assess the causal relationship between 731 immune cell characteristics (divided into seven groups) and osteomyelitis. Genetic variants were used as proxies for risk factors to ensure that the selected instrumental variables meet the three key assumptions of MR analysis. Genome-Wide Association Studies (GWAS) data for immune characteristics were obtained from the public GWAS catalog, while data for osteomyelitis was sourced from the FinnGen. Results At a significance level of 0.05, 21 immune phenotypes were identified as having a causal relationship with osteomyelitis development. In the B cell group, phenotypes such as Memory B cell % B cell (percentage of memory B cells within the total B cell population, % finger cell ratio), CD20- %B cell (percentage of B cells that do not express the CD20 marker on their surface), and Memory B cell % lymphocyte showed a positive causal relationship with osteomyelitis, while Naive-mature B cell %B cell and IgD-CD38-absolute cell counts (AC) phenotypes showed a negative causal relationship. In addition, specific immune phenotypes in the conventional dendritic cells (cDCs) group, Myeloid cell group, TBNK (T cells, B cells, natural killer cells) cell group, T cell maturation stage, and Treg cell group also showed significant associations with osteomyelitis. Through reverse MR analysis, it was found that osteomyelitis had no significant causal impact on these immune phenotypes, suggesting that the occurrence of osteomyelitis may not affect these immune cell phenotypes. Conclusion To our knowledge, this is the first study to shed light on the causal relationship between specific immune cell characteristics and the development of osteomyelitis, thereby providing a new perspective to understand the immune mechanism of osteomyelitis. These findings are significant for formulating targeted prevention and treatment strategies, and hold promise to improve the treatment outcomes for patients with osteomyelitis.
Collapse
Affiliation(s)
- Kehan Long
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ao Gong
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dou Yu
- School of Clinical Medicine, Shandong First Medical University, Jinan, Shandong, China
| | - Sumiao Dong
- School of Clinical Medicine, Shandong First Medical University, Jinan, Shandong, China
| | - Zhendong Ying
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
16
|
Nookaew I, Xiong J, Onal M, Bustamante-Gomez C, Wanchai V, Fu Q, Kim HN, Almeida M, O'Brien CA. Refining the identity of mesenchymal cell types associated with murine periosteal and endosteal bone. J Biol Chem 2024; 300:107158. [PMID: 38479598 PMCID: PMC11007436 DOI: 10.1016/j.jbc.2024.107158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/26/2024] Open
Abstract
Single-cell RNA-seq has led to novel designations for mesenchymal cells associated with bone as well as multiple designations for what appear to be the same cell type. The main goals of this study were to increase the amount of single-cell RNA sequence data for osteoblasts and osteocytes, to compare cells from the periosteum to those inside bone, and to clarify the major categories of cell types associated with murine bone. We created an atlas of murine bone-associated cells by harmonizing published datasets with in-house data from cells targeted by Osx1-Cre and Dmp1-Cre driver strains. Cells from periosteal bone were analyzed separately from those isolated from the endosteum and trabecular bone. Over 100,000 mesenchymal cells were mapped to reveal 11 major clusters designated fibro-1, fibro-2, chondrocytes, articular chondrocytes, tenocytes, adipo-Cxcl12 abundant reticular (CAR), osteo-CAR, preosteoblasts, osteoblasts, osteocytes, and osteo-X, the latter defined in part by periostin expression. Osteo-X, osteo-CAR, and preosteoblasts were closely associated with osteoblasts at the trabecular bone surface. Wnt16 was expressed in multiple cell types from the periosteum but not in cells from endocortical or cancellous bone. Fibro-2 cells, which express markers of stem cells, localized to the periosteum but not trabecular bone in adult mice. Suppressing bone remodeling eliminated osteoblasts and altered gene expression in preosteoblasts but did not change the abundance or location of osteo-X or osteo-CAR cells. These results provide a framework for identifying bone cell types in murine single-cell RNA-seq datasets and suggest that osteoblast progenitors reside near the surface of remodeling bone.
Collapse
Affiliation(s)
- Intawat Nookaew
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Biomedical Informatics, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Jinhu Xiong
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Orthopaedic Surgery, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Melda Onal
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Physiology and Cell Biology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Cecile Bustamante-Gomez
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Visanu Wanchai
- Department of Biomedical Informatics, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Maria Almeida
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Orthopaedic Surgery, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Charles A O'Brien
- Center for Musculoskeletal Disease Research, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Orthopaedic Surgery, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Division of Endocrinology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
17
|
Jackmann N, Gustafsson J, Utriainen P, Magnusson P, Harila A, Atanasova D, Rinaldo C, Frisk P, Mäkitie O. Demographic and disease-related factors impact bone turnover and vitamin D in children with hemato-oncological diseases. JBMR Plus 2024; 8:ziae017. [PMID: 38523666 PMCID: PMC10961176 DOI: 10.1093/jbmrpl/ziae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2024] Open
Abstract
Children with hemato-oncological diseases may have significant skeletal morbidity, not only during and after treatment but also at the time of diagnosis before cancer treatment. This study was designed to evaluate the vitamin D status and circulating bone metabolic markers and their determinants in children at the time of diagnostic evaluation for hemato-oncological disease. This cross-sectional study included 165 children (91 males, median age 6.9 yr range 0.2-17.7 yr). Of them, 76 patients were diagnosed with extracranial or intracranial solid tumors, 83 with leukemia, and 6 with bone marrow failure. Bone metabolism was assessed by measuring serum 25OHD, PTH, bone alkaline phosphatase, intact N-terminal propeptide of type I procollagen, and C-terminal cross-linked telopeptide of type I collagen. Vitamin D deficiency was found in 30.9% of children. Lower 25OHD levels were associated with older age, lack of vitamin D supplementation, season outside summer, and a country of parental origin located between latitudes -45° and 45°. Children diagnosed with leukemia had lower levels of markers of bone formation and bone resorption than those who had solid tumors or bone marrow failure. In conclusion, vitamin D deficiency was observed in one-third of children with newly diagnosed cancer. Bone turnover markers were decreased in children with leukemia, possibly because of the suppression of osteoblasts and osteoclasts by leukemic cells. The identification of patients with suboptimal vitamin D status and compromised bone remodeling at cancer diagnosis may aid in the development of supportive treatment to reduce the adverse effects of cancer and its treatment.
Collapse
Affiliation(s)
- Natalja Jackmann
- Department of Women’s and Children’s Health, Uppsala University and University Children’s Hospital, Uppsala 75185, Sweden
| | - Jan Gustafsson
- Department of Women’s and Children’s Health, Uppsala University and University Children’s Hospital, Uppsala 75185, Sweden
| | - Pauliina Utriainen
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Per Magnusson
- Department of Clinical Chemistry, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58183, Sweden
| | - Arja Harila
- Department of Women’s and Children’s Health, Uppsala University and University Children’s Hospital, Uppsala 75185, Sweden
| | - Diana Atanasova
- Department of Clinical Chemistry, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58183, Sweden
| | - Carina Rinaldo
- Department of Women's and Children's Health, Karolinska Institute, Stockholm 17177, Sweden
| | - Per Frisk
- Department of Women’s and Children’s Health, Uppsala University and University Children’s Hospital, Uppsala 75185, Sweden
| | - Outi Mäkitie
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institute, and Clinical Genetics, Karolinska University Hospital, Stockholm 17177, Sweden
| |
Collapse
|
18
|
Abdulla MA, Hasan RH, Al-Hyani OH. Radiographic and histologic assessment of osseointegration for surface-treated titanium dental implants: An experimental study in dogs. J Dent Res Dent Clin Dent Prospects 2024; 18:44-54. [PMID: 38881643 PMCID: PMC11179146 DOI: 10.34172/joddd.41009] [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: 01/02/2024] [Accepted: 02/23/2024] [Indexed: 06/18/2024] Open
Abstract
Background Modifying the dental implant's surface texture enhances its biological response to the bone and implant‒tissue interface, resulting in the successful support of forces. This study assessed the impact of sandblasting, sandblasting plus acid etching, Er,Cr:YSGG laser, and propolis implant surface treatments and occlusal load on the osseointegration of titanium dental implants in dogs. Methods Seventy-two titanium dental implants with a diameter of 4 mm and a length of 10 mm were divided into four groups according to implant surface modifications (n=18 for each group): group A: sandblasting plus acid etching, group B: sandblasting with Al2 O3, group C: Er,Cr:YSGG laser, group D: propolis coating. Twenty-four local breed male dogs were used. Premolar teeth (P1, P2, and P3) were extracted on the left side of the mandible, and after 12 weeks of bone healing, implants were unilaterally installed. The osseointegration at three study times from implant installation (14, 90, and 180 days) was evaluated. The dog jaws were scanned using an intraoral scanner for the virtual design of screw-retained three-unit crowns after 90 days of osseointegration. Final radiographs were taken before the animals were sacrificed at 14, 90, and 180 days, and the histological analysis was performed. Results Radiographic analysis showed new bone formation (NBF) along and in contact with the implant surface of the treated groups. The histological analysis after 14 days in groups A and B revealed a uniform and ongoing pattern of bone growth and many osteoblasts with few osteocytes within lacunae in new bone trabeculae. Group C showed an increase in the number of osteoblasts lining thin bone trabeculae. Group D showed a generative power concerning bone. At 90 days, there was increased bone ingrowth, and the new bone matured in all the treated implant groups. At 180 days, dense mature bone apposition was in direct contact with delayed-loaded implant surfaces. Conclusion A radiographic examination revealed that surface modification significantly impacted osseointegration, with a strong bond between the implant surface and the surrounding bone. The histological sections at the 14-day interval revealed obvious bone remodeling activity, especially in sandblasting plus acid etching and sandblasting-modified implant surface groups. At the 90-day interval, bone ingrowth had increased, and the new bone became mature, especially in sandblasting and propolis surface modification groups. After 180 days of the delayed-loaded implant osseointegration, differences were observed between different implant-treated groups with a remarkable remodeling of the bone, especially in the propolis coating group.
Collapse
Affiliation(s)
- Mohammed A Abdulla
- Department of Prosthetic Dentistry, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Radhwan H Hasan
- Department of Prosthetic Dentistry, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Osama Hazim Al-Hyani
- Department of Surgery and Theriogenology, College of Veterinary Medicine, University of Mosul, Mosul, Iraq
| |
Collapse
|
19
|
Hegarty-Cremer SGD, Borggaard XG, Andreasen CM, van der Eerden BCJ, Simpson MJ, Andersen TL, Buenzli PR. How osteons form: A quantitative hypothesis-testing analysis of cortical pore filling and wall asymmetry. Bone 2024; 180:116998. [PMID: 38184100 DOI: 10.1016/j.bone.2023.116998] [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: 11/02/2023] [Revised: 12/06/2023] [Accepted: 12/24/2023] [Indexed: 01/08/2024]
Abstract
Osteon morphology provides valuable information about the interplay between different processes involved in bone remodelling. The correct quantitative interpretation of these morphological features is challenging due to the complexity of interactions between osteoblast behaviour, and the evolving geometry of cortical pores during pore closing. We present a combined experimental and mathematical modelling study to provide insights into bone formation mechanisms during cortical bone remodelling based on histological cross-sections of quiescent human osteons and hypothesis-testing analyses. We introduce wall thickness asymmetry as a measure of the local asymmetry of bone formation within an osteon and examine the frequency distribution of wall thickness asymmetry in cortical osteons from human iliac crest bone samples from women 16-78 years old. Our measurements show that most osteons possess some degree of asymmetry, and that the average degree of osteon asymmetry in cortical bone evolves with age. We then propose a comprehensive mathematical model of cortical pore filling that includes osteoblast secretory activity, osteoblast elimination, osteoblast embedment as osteocytes, and osteoblast crowding and redistribution along the bone surface. The mathematical model is first calibrated to symmetric osteon data, and then used to test three mechanisms of asymmetric wall formation against osteon data: (i) delays in the onset of infilling around the cement line; (ii) heterogeneous osteoblastogenesis around the bone perimeter; and (iii) heterogeneous osteoblast secretory rate around the bone perimeter. Our results suggest that wall thickness asymmetry due to off-centred Haversian pores within osteons, and that nonuniform lamellar thicknesses within osteons are important morphological features that can indicate the prevalence of specific asymmetry-generating mechanisms. This has significant implications for the study of disruptions of bone formation as it could indicate what biological bone formation processes may become disrupted with age or disease.
Collapse
Affiliation(s)
- Solene G D Hegarty-Cremer
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane, Australia; Department of Mathematics and Statistics, The University of Montreal, Montreal, Canada
| | - Xenia G Borggaard
- Clinical Cell Biology, Pathology Research Unit, Dept. of Clinical Research, and Dept. of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Dept. of Pathology, Odense University Hospital, Odense, Denmark
| | - Christina M Andreasen
- Clinical Cell Biology, Pathology Research Unit, Dept. of Clinical Research, and Dept. of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Dept. of Pathology, Odense University Hospital, Odense, Denmark
| | | | - Matthew J Simpson
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
| | - Thomas L Andersen
- Clinical Cell Biology, Pathology Research Unit, Dept. of Clinical Research, and Dept. of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Dept. of Pathology, Odense University Hospital, Odense, Denmark; Dept. of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Pascal R Buenzli
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane, Australia.
| |
Collapse
|
20
|
Smit A, Meijer O, Winter E. The multi-faceted nature of age-associated osteoporosis. Bone Rep 2024; 20:101750. [PMID: 38566930 PMCID: PMC10985042 DOI: 10.1016/j.bonr.2024.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Age-associated osteoporosis (AAOP) poses a significant health burden, characterized by increased fracture risk due to declining bone mass and strength. Effective prevention and early treatment strategies are crucial to mitigate the disease burden and the associated healthcare costs. Current therapeutic approaches effectively target the individual contributing factors to AAOP. Nonetheless, the management of AAOP is complicated by the multitude of variables that affect its development. Main intrinsic and extrinsic factors contributing to AAOP risk are reviewed here, including mechanical unloading, nutrient deficiency, hormonal disbalance, disrupted metabolism, cognitive decline, inflammation and circadian disruption. Furthermore, it is discussed how these can be targeted for prevention and treatment. Although valuable as individual targets for intervention, the interconnectedness of these risk factors result in a unique etiology for every patient. Acknowledgement of the multifaceted nature of AAOP will enable the development of more effective and sustainable management strategies, based on a holistic, patient-centered approach.
Collapse
Affiliation(s)
- A.E. Smit
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - O.C. Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - E.M. Winter
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
- Department of Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
21
|
Miller CJ, Trichilo S, Pickering E, Martelli S, Dall'Ara E, Delisser P, Meakin LB, Pivonka P. Cortical thickness adaptation to combined mechanical loading and parathyroid hormone treatments is site specific and synergistic in the mouse tibia model. Bone 2024; 180:116994. [PMID: 38135023 DOI: 10.1016/j.bone.2023.116994] [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: 10/24/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
In this study, we aimed to quantify the localised effects of mechanical loading (ML), low (20 μg/kg/day), moderate (40 μg/kg/day) or high (80 μg/kg/day) dosages of parathyroid hormone (PTH), and combined (PTHML) treatments on cortical bone adaptation in healthy 19-week old female C57BL/6 mice. To this end, we utilise a previously reported image analysis algorithm on μCT data of the mouse tibia published by Sugiyama et al. (2008) to measure changes in cortical area, marrow cavity area and local cortical thickness measures (ΔCt.Ar, ΔMa.Ar, ΔCt.Th respectively), evaluated at two cross-sections within the mouse tibia (proximal-middle (37 %) and middle (50 %)), and are compared to a superposed summation (P + M) of individual treatments to determine the effectiveness of combining treatments in vivo. ΔCt.Ar analysis revealed a non-linear, synergistic interactions between PTH and ML in the 37 % cross-section that saturates at higher PTH dosages, whereas the 50 % cross-section experiences an approximately linear, additive adaptation response. This coincided with an increase in ΔMa.Ar (indicating resorption of the endosteal surface), which was only counteracted by combined high dose PTH with ML in the middle cross-section. Regional analysis of ΔCt.Th changes reveal localised cortical thinning in response to low dose PTH treatment in the posteromedial region of the middle cross-section, signifying that PTH does not provide a homogeneous adaptation response around the cortical perimeter. We observe a synergistic response in the proximal-middle cross-section, with regions of compressive strain experiencing the greatest adaptation response to PTHML treatments, (peak ΔCt.Th of 189.32, 213.78 and 239.30 μm for low, moderate and high PTHML groups respectively). In contrast, PTHML treatments in the middle cross-section show a similar response to the superposed P + M group, with the exception of the combined high dose PTHML treatment which shows a synergistic interaction. These analyses suggest that, in mice, adding mechanical loading to PTH treatments leads to region specific bone responses; synergism of PTHML is only achieved in some regions experiencing high loading, while other regions respond additively to this combined treatment.
Collapse
Affiliation(s)
- Corey J Miller
- Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Edmund Pickering
- Queensland University of Technology, Brisbane, Queensland, Australia
| | - Saulo Martelli
- Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Peter Delisser
- Veterinary Specialist Services, Brisbane, Queensland, Australia
| | | | - Peter Pivonka
- Queensland University of Technology, Brisbane, Queensland, Australia.
| |
Collapse
|
22
|
Kim K, Kim JH, Kim I, Seong S, Koh JT, Kim N. Sestrin2 inhibits RANKL-induced osteoclastogenesis through AMPK activation and ROS inhibition. Free Radic Biol Med 2024; 211:77-88. [PMID: 38101586 DOI: 10.1016/j.freeradbiomed.2023.12.009] [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: 09/12/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
Sestrins are stress-responsive proteins with antioxidant properties. They participate in cellular redox balance and protect against oxidative damage. This study investigated the effects of Sestrin2 (Sesn2) on osteoclast differentiation and function. Overexpressing Sesn2 in osteoclast precursor cells significantly inhibited receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis. This was assessed as reduced expression of various osteoclast markers, including c-Fos, nuclear factor of activated T cells 1 (NFATc1), osteoclast-associated receptor, tartrate-resistant acid phosphatase, and cathepsin K. Conversely, downregulation of Sesn2 produced the opposite effect. Mechanistically, Sesn2 overexpression enhanced AMPK activation and the nuclear translocation of nuclear factor erythroid-derived factor 2-related factor 2 (Nrf2), promoting antioxidant enzymes. Moreover, azithromycin (Azm) induced Sesn2 expression, which suppressed RANKL-induced osteoclast differentiation. Specifically, Azm treatment reduced RANKL-induced production of reactive oxygen species in osteoclasts. Furthermore, intraperitoneal administration of Azm ameliorated RANKL-induced bone loss by reducing osteoclast activity in mice. Taken together, our results suggested that Azm-induced Sesn2 act as a negative regulator of RANKL-induced osteoclast differentiation through the AMPK/NFATc1 signaling pathway. Concisely, targeting Sesn2 can be a potential pharmacological intervention in osteoporosis.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
23
|
Cao Z, Qin Z, Duns GJ, Huang Z, Chen Y, Wang S, Deng R, Nie L, Luo X. Repair of Infected Bone Defects with Hydrogel Materials. Polymers (Basel) 2024; 16:281. [PMID: 38276689 PMCID: PMC10820481 DOI: 10.3390/polym16020281] [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: 12/13/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Infected bone defects represent a common clinical condition involving bone tissue, often necessitating surgical intervention and antibiotic therapy. However, conventional treatment methods face obstacles such as antibiotic resistance and susceptibility to postoperative infections. Hydrogels show great potential for application in the field of tissue engineering due to their advantageous biocompatibility, unique mechanical properties, exceptional processability, and degradability. Recent interest has surged in employing hydrogels as a novel therapeutic intervention for infected bone repair. This article aims to comprehensively review the existing literature on the anti-microbial and osteogenic approaches utilized by hydrogels in repairing infected bones, encompassing their fabrication techniques, biocompatibility, antimicrobial efficacy, and biological activities. Additionally, the potential opportunities and obstacles in their practical implementation will be explored. Lastly, the limitations presently encountered and the prospective avenues for further investigation in the realm of hydrogel materials for the management of infected bone defects will be deliberated. This review provides a theoretical foundation and advanced design strategies for the application of hydrogel materials in the treatment of infected bone defects.
Collapse
Affiliation(s)
- Zhenmin Cao
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Zuodong Qin
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Gregory J. Duns
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Zhao Huang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Yao Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Sheng Wang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Ruqi Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Libo Nie
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Xiaofang Luo
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| |
Collapse
|
24
|
Silva AV, Gomes DDS, Victor RDS, Santana LNDL, Neves GA, Menezes RR. Influence of Strontium on the Biological Behavior of Bioactive Glasses for Bone Regeneration. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7654. [PMID: 38138796 PMCID: PMC10744628 DOI: 10.3390/ma16247654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Bioactive glasses (BGs) can potentially be applied in biomedicine, mainly for bone repair and replacement, given their unique ability to connect to natural bone tissue and stimulate bone regeneration. Since their discovery, several glass compositions have been developed to improve the properties and clinical abilities of traditional bioactive glass. Different inorganic ions, such as strontium (Sr2+), have been incorporated in BG due to their ability to perform therapeutic functions. Sr2+ has been gaining prominence due to its ability to stimulate osteogenesis, providing an appropriate environment to improve bone regeneration, in addition to its antibacterial potential. However, as there are still points in the literature that are not well consolidated, such as the influence of ionic concentrations and the BG production technique, this review aims to collect information on the state of the art of the biological behavior of BGs containing Sr2+. It also aims to gather data on different types of BGs doped with different concentrations of Sr2+, and to highlight the manufacturing techniques used in order to analyze the influence of the incorporation of this ion for bone regeneration purposes.
Collapse
Affiliation(s)
- Amanda Vieira Silva
- Graduate Program in Materials Science and Engineering, Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil;
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| | - Déborah dos Santos Gomes
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| | - Rayssa de Sousa Victor
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| | - Lisiane Navarro de Lima Santana
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| | - Gelmires Araújo Neves
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| | - Romualdo Rodrigues Menezes
- Laboratory of Materials Technology (LTM), Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil; (R.d.S.V.); (L.N.d.L.S.); (G.A.N.)
| |
Collapse
|
25
|
Faienza MF, Urbano F, Chiarito M, Lassandro G, Giordano P. Musculoskeletal health in children and adolescents. Front Pediatr 2023; 11:1226524. [PMID: 38161439 PMCID: PMC10754974 DOI: 10.3389/fped.2023.1226524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
The purpose of this narrative review was to investigate the key determinants of musculoskeletal health in childhood and adolescence, with particular attention to the role of physical activity. First, we examined the importance of bone modeling and remodeling in maintaining the bone health and the integrity and mechanical characteristic of the skeleton. In addition, we reported the evidence on an appropriate calcium and vitamin D intake, as well as local load variation in achieving proper peak bone mass. Proteomic and transcriptomic studies identified the skeletal muscle "secretoma", consisting of several myokines involved in endocrine and paracrine functions. Among these, we explored the role of irisin, a myokine involved in the muscle-bone crosstalk, and in the regulation of metabolic pathways. It is known that physical activity during growing positively impacts on skeleton and can protect by bone loss in adulthood. However, there are still concerns about the optimal interval duration and exercise intensity, particularly at the pubertal growth spurt which represents a window of opportunity to increase skeletal strength. We reported data from clinical trials performed in the last 5 years analyzing the impact of the type and timing of physical activity during childhood on skeletal development. Finally, we reported recent data on the significance of physical activity in some rare diseases.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Paola Giordano
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
26
|
Karkache IY, Molstad DHH, Vu E, Jensen ED, Bradley EW. Phlpp1 Expression in Osteoblasts Plays a Modest Role in Bone Homeostasis. JBMR Plus 2023; 7:e10806. [PMID: 38130760 PMCID: PMC10731110 DOI: 10.1002/jbm4.10806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 12/23/2023] Open
Abstract
Prior work demonstrated that Phlpp1 deficiency alters limb length and bone mass, but the cell types involved and requirement of Phlpp1 for this effect were unclear. To understand the function of Phlpp1 within bone-forming osteoblasts, we crossed Phlpp1 floxed mice with mice harboring type 1 collagen (Col1a12.3kb)-Cre. Mineralization of bone marrow stromal cell cultures derived from Phlpp1 cKOCol1a1 was unchanged, but levels of inflammatory genes (eg, Ifng, Il6, Ccl8) and receptor activator of NF-κB ligand/osteoprotegerin (RANKL/OPG) ratios were enhanced by either Phlpp1 ablation or chemical inhibition. Micro-computed tomography of the distal femur and L5 vertebral body of 12-week-old mice revealed no alteration in bone volume per total volume, but compromised femoral bone microarchitecture within Phlpp1 cKOCol1a1 conditional knockout females. Bone histomorphometry of the proximal tibia documented no changes in osteoblast or osteoclast number per bone surface but slight reductions in osteoclast surface per bone surface. Overall, our data show that deletion of Phlpp1 in type 1 collagen-expressing cells does not significantly alter attainment of peak bone mass of either males or females, but may enhance inflammatory gene expression and the ratio of RANKL/OPG. Future studies examining the role of Phlpp1 within models of advanced age, inflammation, or osteocytes, as well as functional redundancy with the related Phlpp2 isoform are warranted. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ismael Y Karkache
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
- College of Veterinary SciencesUniversity of MinnesotaMinneapolisMNUSA
| | - David HH Molstad
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
| | - Elizabeth Vu
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
| | | | - Elizabeth W Bradley
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
- College of Veterinary SciencesUniversity of MinnesotaMinneapolisMNUSA
- Department of Orthopedic SurgeryStem Cell Institute, University of MinnesotaMinneapolisMNUSA
| |
Collapse
|
27
|
van Dijk Christiansen P, Andreasen CM, El-Masri BM, Laursen KS, Delaisse JM, Andersen TL. Osteoprogenitor recruitment and differentiation during intracortical bone remodeling of adolescent humans. Bone 2023; 177:116896. [PMID: 37699496 DOI: 10.1016/j.bone.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Recruitment and proliferation of osteoprogenitors during the reversal-resorption phase, and their differentiation into mature bone-forming osteoblasts is crucial for initiation of bone formation during bone remodeling. This study investigates the osteoprogenitors' gradual recruitment, proliferation, and differentiation into bone-forming osteoblasts within intracortical remodeling events of healthy adolescent humans. METHODS The study was conducted on cortical bone specimens from 11 adolescent human controls - patients undergoing surgery due to coxa valga. The osteoprogenitor recruitment route and differentiation into osteoblasts were backtracked using immunostainings and in situ hybridizations with osteoblastic markers (CD271/NGFR, osterix/SP7, COL3A1 and COL1A1). The osteoblastic cell populations were defined based on the pore surfaces, and their proliferation index (Ki67), density and number/circumference were estimated in multiplex-immunofluorescence (Ki67, TRAcP, CD34) stained sections. RESULTS During the reversal-resorption phase, osteoclasts are intermixed with (COL3A1+NFGR+) osteoblastic reversal cells, which are considered to be osteoprogenitors of (COL1A1+SP7+) bone-forming osteoblasts. Initiation of bone formation requires a critical density of these osteoprogenitors (43 ± 9 cells/mm), which is reached though proliferation (4.4 ± 0.5 % proliferative) and even more so through recruitment of osteoprogenitors, but challenged by the ongoing expansion of the canal circumference. These osteoprogenitors most likely originate from osteoblastic bone lining cells and mainly lumen osteoprogenitors, which expand their population though proliferation (4.6 ± 0.3 %) and vascular recruitment. These lumen osteoprogenitors resemble canopy cells above trabecular remodeling sites, and like canopy cells they extend above bone-forming osteoblasts where they may rejuvenate the osteoblast population during bone formation. CONCLUSION Initiation of bone formation during intracortical remodeling requires a critical density of osteoprogenitors on eroded surfaces, which is reached though proliferation and recruitment of local osteoprogenitors: bone lining cells and lumen osteoprogenitors.
Collapse
Affiliation(s)
- Pernille van Dijk Christiansen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Christina Møller Andreasen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Bilal Mohamad El-Masri
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Kaja Søndergaard Laursen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Thomas Levin Andersen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Forensic Medicine, Aarhus University, Aarhus, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| |
Collapse
|
28
|
Nookaew I, Xiong J, Onal M, Bustamante-Gomez C, Wanchai V, Fu Q, Kim HN, Almeida M, O'Brien CA. A framework for defining mesenchymal cell types associated with murine periosteal and endosteal bone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567528. [PMID: 38014179 PMCID: PMC10680810 DOI: 10.1101/2023.11.17.567528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Single-cell RNA sequencing has led to numerous novel designations for mesenchymal cell types associated with bone. Consequently, there are now multiple designations for what appear to be the same cell type. In addition, existing datasets contain relatively small numbers of mature osteoblasts and osteocytes and there has been no comparison of periosteal bone cells to those at the endosteum and trabecular bone. The main goals of this study were to increase the amount of single cell RNA sequence data for osteoblasts and osteocytes, to compare cells from the periosteum to those inside bone, and to clarify the major categories of cell types associated with murine bone. To do this, we created an atlas of murine bone-associated cells by harmonizing published datasets with in-house data from cells targeted by Osx1-Cre and Dmp1-Cre driver strains. Cells from periosteal bone were analyzed separately from those isolated from the endosteum and trabecular bone. Over 100,000 mesenchymal cells were mapped to reveal 11 major clusters designated fibro-1, fibro-2, chondrocytes, articular chondrocytes, tenocytes, adipo-CAR, osteo-CAR, pre-osteoblasts, osteoblasts, osteocytes, and osteo-X, the latter defined in part by Postn expression. Osteo-X, osteo-CAR, and pre-osteoblasts were closely associated with osteoblasts at the trabecular bone surface. Wnt16 was expressed in multiple cell types from the periosteum but not in any cells from endocortical or cancellous bone. Fibro-2 cells, which express markers of skeletal stem cells, localized to the periosteum but not trabecular bone in adult mice. Suppressing bone remodeling eliminated osteoblasts and altered gene expression in pre-osteoblasts but did not change the abundance or location of osteo-X or osteo-CAR cells. These results provide a framework for identifying bone cell types in murine single cell RNA sequencing datasets and suggest that osteoblast progenitors reside near the surface of remodeling bone.
Collapse
Affiliation(s)
- Intawat Nookaew
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Biomedical Informatics, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jinhu Xiong
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Orthopaedic Surgery, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Melda Onal
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Physiology and Cell Biology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Cecile Bustamante-Gomez
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Division of Endocrinology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Visanu Wanchai
- Department of Biomedical Informatics, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Qiang Fu
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Division of Endocrinology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Division of Endocrinology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Maria Almeida
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Orthopaedic Surgery, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Division of Endocrinology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Charles A O'Brien
- Center for Musculoskeletal Disease Research, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Orthopaedic Surgery, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Division of Endocrinology, the University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
| |
Collapse
|
29
|
Bai Y, Bentley L, Ma C, Naveenan N, Cleak J, Wu Y, Simon MM, Westerberg H, Cañas RC, Horner N, Pandey R, Paphiti K, Schulze U, Mianné J, Hough T, Teboul L, de Baaij JH, Cox RD. Cleft palate and minor metabolic disturbances in a mouse global Arl15 gene knockout. FASEB J 2023; 37:e23211. [PMID: 37773757 PMCID: PMC10631251 DOI: 10.1096/fj.202201918r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 07/27/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023]
Abstract
ARL15, a small GTPase protein, was linked to metabolic traits in association studies. We aimed to test the Arl15 gene as a functional candidate for metabolic traits in the mouse. CRISPR/Cas9 germline knockout (KO) of Arl15 showed that homozygotes were postnatal lethal and exhibited a complete cleft palate (CP). Also, decreased cell migration was observed from Arl15 KO mouse embryonic fibroblasts (MEFs). Metabolic phenotyping of heterozygotes showed that females had reduced fat mass on a chow diet from 14 weeks of age. Mild body composition phenotypes were also observed in heterozygous mice on a high-fat diet (HFD)/low-fat diet (LFD). Females on a HFD showed reduced body weight, gonadal fat depot weight and brown adipose tissue (BAT) weight. In contrast, in the LFD group, females showed increased bone mineral density (BMD), while males showed a trend toward reduced BMD. Clinical biochemistry analysis of plasma on HFD showed transient lower adiponectin at 20 weeks of age in females. Urinary and plasma Mg2+ concentrations were not significantly different. Our phenotyping data showed that Arl15 is essential for craniofacial development. Adult metabolic phenotyping revealed potential roles in brown adipose tissue and bone development.
Collapse
Affiliation(s)
- Ying Bai
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Liz Bentley
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Chao Ma
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - James Cleak
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Yixing Wu
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Michelle M Simon
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Henrik Westerberg
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Ramón Casero Cañas
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Neil Horner
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Rajesh Pandey
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Keanu Paphiti
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | | | - Joffrey Mianné
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Tertius Hough
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Lydia Teboul
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| | - Jeroen H.F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Roger D. Cox
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Didcot, Oxon OX11 0RD, UK
| |
Collapse
|
30
|
Whitney DG, Caird MS, Raggio CL, Hurvitz EA, Clines GA, Jepsen KJ. Perspective: A multi-trait integrative approach to understanding the structural basis of bone fragility for pediatric conditions associated with abnormal bone development. Bone 2023; 175:116855. [PMID: 37481149 DOI: 10.1016/j.bone.2023.116855] [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: 06/08/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Bone development is a highly orchestrated process that establishes the structural basis of bone strength during growth and functionality across the lifespan. This developmental process is generally robust in establishing mechanical function, being adaptable to many genetic and environmental factors. However, not all factors can be fully accommodated, leading to abnormal bone development and lower bone strength. This can give rise to early-onset bone fragility that negatively impacts bone strength across the lifespan. Current guidelines for assessing bone strength include measuring bone mineral density, but this does not capture the structural details responsible for whole bone strength in abnormally developing bones that would be needed to inform clinicians on how and when to treat to improve bone strength. The clinical consequence of not operationalizing how altered bone development informs decision making includes under-detection and missed opportunities for early intervention, as well as a false positive diagnosis of fragility with possible resultant clinical actions that may actually harm the growing skeleton. In this Perspective, we emphasize the need for a multi-trait, integrative approach to better understand the structural basis of bone growth for pediatric conditions with abnormal bone development. We provide evidence to showcase how this approach might reveal multiple, unique ways in which bone fragility develops across and within an array of pediatric conditions that are associated with abnormal bone development. This Perspective advocates for the development of new translational research aimed at informing better ways to optimize bone growth, prevent fragility fractures, and monitor and treat bone fragility based on the child's skeletal needs.
Collapse
Affiliation(s)
- Daniel G Whitney
- Department of Physical Medicine and Rehabilitation, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA.
| | - Michelle S Caird
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Edward A Hurvitz
- Department of Physical Medicine and Rehabilitation, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gregory A Clines
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Endocrinology Section, Ann Arbor VA Medical Center, Ann Arbor, MI, USA
| | - Karl J Jepsen
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
31
|
de Wildt BWM, Cuypers LAB, Cramer EEA, Wentzel AS, Ito K, Hofmann S. The Impact of Culture Variables on a 3D Human In Vitro Bone Remodeling Model: A Design of Experiments Approach. Adv Healthc Mater 2023; 12:e2301205. [PMID: 37405830 PMCID: PMC11469142 DOI: 10.1002/adhm.202301205] [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: 04/17/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
Human in vitro bone remodeling models, using osteoclast-osteoblast cocultures, can facilitate the investigation of human bone remodeling while reducing the need for animal experiments. Although current in vitro osteoclast-osteoblast cocultures have improved the understanding of bone remodeling, it is still unknown which culture conditions support both cell types. Therefore, in vitro bone remodeling models can benefit from a thorough evaluation of the impact of culture variables on bone turnover outcomes, with the aim to reach balanced osteoclast and osteoblast activity, mimicking healthy bone remodeling. Using a resolution III fractional factorial design, the main effects of commonly used culture variables on bone turnover markers in an in vitro human bone remodeling model are identified. This model is able to capture physiological quantitative resorption-formation coupling along all conditions. Culture conditions of two runs show promising results: conditions of one run can be used as a high bone turnover system and conditions of another run as a self-regulating system as the addition of osteoclastic and osteogenic differentiation factors is not required for remodeling. The results generated with this in vitro model allow for better translation between in vitro studies and in vivo studies, toward improved preclinical bone remodeling drug development.
Collapse
Affiliation(s)
- Bregje W. M. de Wildt
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Lizzy A. B. Cuypers
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Regenerative BiomaterialsRadboud Institute for Molecular Life SciencesRadboud University Medical CenterP.O. Box 9101Nijmegen6525 GAThe Netherlands
| | - Esther E. A. Cramer
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Annelieke S. Wentzel
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics and Institute for Complex Molecular Systems (ICMS)Department of Biomedical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| |
Collapse
|
32
|
Jeong H, Kim D, Montagne K, Ushida T, Furukawa KS. Differentiation-inducing effect of osteoclast microgrooves for the purpose of three-dimensional design of regenerated bone. Acta Biomater 2023; 168:174-184. [PMID: 37392936 DOI: 10.1016/j.actbio.2023.06.033] [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: 04/24/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
In vivo bone remodeling is promoted by the balance between osteoclast and osteoblast activity. Conventional research on bone regeneration has mainly focused on increasing osteoblast activity, with limited studies on the effects of scaffold topography on cell differentiation. Here, we examined the effect of microgroove-patterned substrate with spacings ranging from 1 to 10 μm on the differentiation of rat bone marrow-derived osteoclast precursors. Tartrate-resistant acid phosphatase (TRAP) staining and relative gene expression quantification showed that osteoclast differentiation was enhanced in substrate with 1 µm microgroove spacing compared with that in the other groups. Additionally, the ratio of podosome maturation stages in substrate with 1 μm microgroove spacing exhibited a distinct pattern, which was characterized by an increase in the ratio of belts and rings and a decrease in that of clusters. However, myosin II abolished the effects of topography on osteoclast differentiation. Overall, these showed that the reduction of myosin II tension in the podosome core by an integrin vertical vector increased podosome stability and promoted osteoclast differentiation in substrates with 1 μm microgroove spacing, including that microgroove design plays an important role in scaffolds for bone regeneration. STATEMENT OF SIGNIFICANCE: Reduction of myosin II tension in the podosome core, facilitated by an integrin vertical vector, resulted in an enhanced osteoclast differentiation, concomitant with an increase in podosome stability within 1-μm-spaced microgrooves. These findings are anticipated to serve as valuable indicators for the regulation of osteoclast differentiation through the manipulation of biomaterial surface topography in tissue engineering. Furthermore, this study contributes to the lucidation of the underlying mechanisms governing cellular differentiation by providing insights into the impact of the microtopographical environment.
Collapse
Affiliation(s)
- Heonuk Jeong
- Department of Bioengineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Dain Kim
- Department of Mechanical Engineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Kevin Montagne
- Department of Mechanical Engineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Takashi Ushida
- Department of Mechanical Engineering, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Katsuko S Furukawa
- Department of Bioengineering, School of Engineering, University of Tokyo, Tokyo, Japan; Department of Mechanical Engineering, School of Engineering, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
33
|
Chen X, Chen X, Chao R, Wang Y, Mao Y, Fan B, Zhang Y, Xu W, Qin A, Zhang S. Dlk2 interacts with Syap1 to activate Akt signaling pathway during osteoclast formation. Cell Death Dis 2023; 14:589. [PMID: 37669921 PMCID: PMC10480461 DOI: 10.1038/s41419-023-06107-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023]
Abstract
Excessive osteoclast formation and bone resorption are related to osteolytic diseases. Delta drosophila homolog-like 2 (Dlk2), a member of the epidermal growth factor (EGF)-like superfamily, reportedly regulates adipocyte differentiation, but its roles in bone homeostasis are unclear. In this study, we demonstrated that Dlk2 deletion in osteoclasts significantly inhibited osteoclast formation in vitro and contributed to a high-bone-mass phenotype in vivo. Importantly, Dlk2 was shown to interact with synapse-associated protein 1 (Syap1), which regulates Akt phosphorylation at Ser473. Dlk2 deletion inhibited Syap1-mediated activation of the AktSer473, ERK1/2 and p38 signaling cascades. Additionally, Dlk2 deficiency exhibits increased bone mass in ovariectomized mice. Our results reveal the important roles of the Dlk2-Syap1 signaling pathway in osteoclast differentiation and osteoclast-related bone disorders.
Collapse
Affiliation(s)
- Xinwei Chen
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Xuzhuo Chen
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Rui Chao
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Yexin Wang
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Yi Mao
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Baoting Fan
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Yaosheng Zhang
- Department of Stomatology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Weifeng Xu
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China.
| | - An Qin
- Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai, People's Republic of China.
| | - Shanyong Zhang
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China.
| |
Collapse
|
34
|
Cai G, Lu Y, Zhong W, Wang T, Li Y, Ruan X, Chen H, Sun L, Guan Z, Li G, Zhang H, Sun W, Chen M, Zhang W, Wang H. Piezo1-mediated M2 macrophage mechanotransduction enhances bone formation through secretion and activation of transforming growth factor-β1. Cell Prolif 2023; 56:e13440. [PMID: 36880296 PMCID: PMC10472522 DOI: 10.1111/cpr.13440] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/02/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Macrophages are multifunctional immune system cells that are essential for the mechanical stimulation-induced control of metabolism. Piezo1 is a non-selective calcium channel expressed in multifarious tissues to convey mechanical signals. Here, a cellular model of tension was used to study the effect of mechanical stretch on the phenotypic transformation of macrophages and its mechanism. An indirect co-culture system was used to explore the effect of macrophage activation on bone marrow mesenchymal stem cells (BMSCs), and a treadmill running model was used to validate the mechanism in vivo for in vitro studies. p53 was acetylated and deacetylated by macrophages as a result of mechanical strain being detected by Piezo1. This process is able to polarize macrophages towards M2 and secretes transforming growth factor-beta (TGF-β1), which subsequently stimulates BMSCs migration, proliferation and osteogenic differentiation. Knockdown of Piezo1 inhibits the conversion of macrophages to the reparative phenotype, thereby affecting bone remodelling. Blockade of TGF-β I, II receptors and Piezo1 significantly reduced exercise-increased bone mass in mice. In conclusion, we showed that mechanical tension causes calcium influx, p53 deacetylation, macrophage polarization towards M2 and TGF-β1 release through Piezo1. These events support BMSC osteogenesis.
Collapse
Affiliation(s)
- Guanhui Cai
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Yahui Lu
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingChina
| | - Weijie Zhong
- Department of StomatologyDushu Lake Hospital Affiliated to Soochow UniversitySoochowChina
- Department of StomatologyMedical Center of Soochow UniversitySoochowChina
| | - Ting Wang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Yingyi Li
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiaolei Ruan
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Hongyu Chen
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Lian Sun
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhaolan Guan
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Gen Li
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Hengwei Zhang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Wen Sun
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Minglong Chen
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Wei‐Bing Zhang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- Department of StomatologyDushu Lake Hospital Affiliated to Soochow UniversitySoochowChina
- Department of StomatologyMedical Center of Soochow UniversitySoochowChina
| | - Hua Wang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of OrthodonticsJiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| |
Collapse
|
35
|
Maisuria R, Norton A, Shao C, Bradley EW, Mansky K. Conditional Loss of MEF2C Expression in Osteoclasts Leads to a Sex-Specific Osteopenic Phenotype. Int J Mol Sci 2023; 24:12686. [PMID: 37628864 PMCID: PMC10454686 DOI: 10.3390/ijms241612686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Myocyte enhancement factor 2C (MEF2C) is a transcription factor studied in the development of skeletal and smooth muscles. Bone resorption studies have exhibited that the reduced expression of MEF2C contributes to osteopetrosis and the dysregulation of pathological bone remodeling. Our current study aims to determine how MEF2C contributes to osteoclast differentiation and to analyze the skeletal phenotype of Mef2c-cKO mice (Cfms-cre; Mef2cfl/fl). qRT-PCR and Western blot demonstrated that Mef2c expression is highest during the early days of osteoclast differentiation. Osteoclast genes, including c-Fos, c-Jun, Dc-stamp, Cathepsin K, and Nfatc1, had a significant reduction in expression, along with a reduction in osteoclast size. Despite reduced CTX activity, female Mef2c cKO mice were osteopenic, with decreased bone formation as determined via a P1NP ELISA, and a reduced number of osteoblasts. There was no difference between male WT and Mef2c-cKO mice. Our results suggest that Mef2c is critical for osteoclastogenesis, and that its dysregulation leads to a sex-specific osteopenic phenotype.
Collapse
Affiliation(s)
- Ravi Maisuria
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Andrew Norton
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Cynthia Shao
- College of Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Elizabeth W. Bradley
- Department of Orthopedics, School of Medicine and Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Kim Mansky
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| |
Collapse
|
36
|
Chen X, GUO Y, LU J, QIN L, HU T, ZENG X, WANG X, ZHANG A, ZHUANG Y, ZHONG H, GUO C. Acupotomy ameliorates subchondral bone absorption and mechanical properties in rabbits with knee osteoarthritis by regulating bone morphogenetic protein 2-Smad1 pathway. J TRADIT CHIN MED 2023; 43:734-743. [PMID: 37454258 PMCID: PMC10320461 DOI: 10.19852/j.cnki.jtcm.20230404.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/13/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE To investigate the effects of acupotomy on the subchondral bone absorption and mechanical properties in rabbits with knee osteoarthritis (KOA). METHODS The rabbits were divided into blank control, model, acupotomy and electroacupuncture (EA) groups, with 12 rabbits in each. Modified Videman's method was used to prepare KOA model. The acupotomy and EA group were given indicated intervention for 3 weeks. The behavior of rabbits in each group was recorded. Subsequently, cartilage-subchondral bone units were obtained and morphological changes were observed by optical microscope and micro computed tomography. Compression test was used to detect the mechanical properties of subchondral bone, Western blot and real-time polymerase chain reaction (RT-PCR) were applied to detect the expression of bone morphogenetic protein 2-Smad1 (BMP2-Smad1) pathway in subchondral bone. RESULTS Compared with the control group, rabbits in the KOA group showed lameness, knee pain, and cartilage degradation; the subchondral bone showed active resorption, the mechanical properties decreased significantly and the BMP2-Smad1 pathway downregulated significantly. Both acupotomy and EA intervention could increase the thickness of trabecular bone (Tb. Th), the bone volume fraction (BV/TV) and the thickness of subchondral bone plate, reduce the separation of trabecular bone (Tb. Sp), improve the maximum load and elastic modulus of subchondral bone, and effectively delay cartilage degeneration in KOA rabbits. This process may be achieved through upregulation the related proteins of BMP2-Smad1 pathway. The maximum load and elastic modulus of subchondral bone in the acupotomy group were slightly better than those in the EA group. CONCLUSIONS Acupotomy could effectively protect cartilage by inhibiting abnormal bone resorption and improving mechanical properties of subchondral bone thorough the related proteins of BMP2-Smad1 pathway in KOA rabbits.
Collapse
Affiliation(s)
- Xilin Chen
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan GUO
- 2 Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated with Capital Medical University, Beijing 100010, China
| | - Juan LU
- 3 Shenzhen Hospital of Southern Medical University, Shenzhen 518000, China
| | - Luxue QIN
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tingyao HU
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xin ZENG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinyue WANG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Anran ZHANG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuxin ZHUANG
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Honggang ZHONG
- 4 Institute of Bone Injury, China Academy of Chinese Medical Sciences, Beijing 100010, China
| | - Changqing GUO
- 1 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
37
|
Pan L, Zhang C, Zhang H, Ke T, Bian M, Yang Y, Chen L, Tan J. Osteoclast-Derived Exosomal miR-5134-5p Interferes with Alveolar Bone Homeostasis by Targeting the JAK2/STAT3 Axis. Int J Nanomedicine 2023; 18:3727-3744. [PMID: 37441084 PMCID: PMC10335290 DOI: 10.2147/ijn.s413692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Background In chronic periodontitis, exosomes transport various informative substances between osteoclasts and osteoblasts in alveolar bone. Herein, we aimed to investigate the effect of exosomal micro-ribonucleic acid (miRNA/miR)-5134-5p derived from osteoclasts on osteoblastic proliferation and differentiation and the development of periodontitis in vivo and in vitro. Methods The effects of OC-Exos on the proliferation and differentiation of osteoblasts were identified by Real-time quantitative reverse polymerase chain reaction (qRT-PCR), Western blot(WB), alkaline phosphatase(ALP) staining, etc. Exosomal miRNA expression was analyzed by sequencing. The sites of miRNA action were predicted through TargetScan and tested by double luciferase assay. After transfecting miR-5134-5p mimic/inhibitor into osteoblasts, we measured the proliferation and differentiation of osteoblasts by ALP staining and WB, etc. Furthermore, OC-Exos were injected into the gingival sulcus at the ligation site. Inflammation was observed by Hematoxylin-eosin (H&E) staining, the expression of inflammatory factors were detected by qRT-PCR, the resorption of alveolar bone was observed by Micro CT. Results Osteoblastic proliferation and differentiation were negatively regulated by OC-Exos in vitro. miRNA sequencing analysis revealed that miR-5134-5p expression was significantly elevated in OC-Exos, which also increased in osteoblasts following OC-Exo intervention. The dual-luciferase assay revealed that miR-5134-5p and Janus kinase 2 (JAK2) had binding sites. miR-5134-5p-mimics could upregulate miR-5134-5p expression in osteoblasts while downregulating Runt-related transcription factor 2(Runx2), phosphorylated-JAK2 (p-JAK2), and phosphorylated-signal transducer and activator of transcription 3 (p-STAT3) expression and inhibited osteogenic differentiation. However, miR-5134-5p-inhibitor had the opposite effect. In vivo, the OC-Exo group demonstrated morphological disruption of periodontal tissue, massive inflammatory cell infiltration, upregulation of inflammatory factors mRNA expression, a significant decrease in BV/TV, and an increase in the cementoenamel junction and alveolar bone crest distance. Conclusion Osteoclast-derived exosomal miR-5134-5p inhibits osteoblastic proliferation and differentiation via the JAK2/STAT3 pathway. OC-Exos exacerbate periodontal tissue inflammation and accelerate alveolar bone resorption in mice with experimental periodontitis.
Collapse
Affiliation(s)
- Lai Pan
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Chenyi Zhang
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Haizheng Zhang
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Ting Ke
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Mengyao Bian
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Yuxuan Yang
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Lili Chen
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Jingyi Tan
- Department of Periodontology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, People’s Republic of China
| |
Collapse
|
38
|
Bonanni R, Cariati I, Marini M, Tarantino U, Tancredi V. Microgravity and Musculoskeletal Health: What Strategies Should Be Used for a Great Challenge? Life (Basel) 2023; 13:1423. [PMID: 37511798 PMCID: PMC10381503 DOI: 10.3390/life13071423] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Space colonization represents the most insidious challenge for mankind, as numerous obstacles affect the success of space missions. Specifically, the absence of gravitational forces leads to systemic physiological alterations, with particular emphasis on the musculoskeletal system. Indeed, astronauts exposed to spaceflight are known to report a significant impairment of bone microarchitecture and muscle mass, conditions clinically defined as osteoporosis and sarcopenia. In this context, space medicine assumes a crucial position, as the development of strategies to prevent and/or counteract weightlessness-induced alterations appears to be necessary. Furthermore, the opportunity to study the biological effects induced by weightlessness could provide valuable information regarding adaptations to spaceflight and suggest potential treatments that can preserve musculoskeletal health under microgravity conditions. Noteworthy, improving knowledge about the latest scientific findings in this field of research is crucial, as is thoroughly investigating the mechanisms underlying biological adaptations to microgravity and searching for innovative solutions to counter spaceflight-induced damage. Therefore, this narrative study review, performed using the MEDLINE and Google Scholar databases, aims to summarize the most recent evidence regarding the effects of real and simulated microgravity on the musculoskeletal system and to discuss the effectiveness of the main defence strategies used in both real and experimental settings.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Clinical Sciences and Translational Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Mario Marini
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
39
|
Sauvé B, Chorfi Y, Montminy MPL, Guay F. Vitamin D Supplementation Impacts Calcium and Phosphorus Metabolism in Piglets Fed a Diet Contaminated with Deoxynivalenol and Challenged with Lipopolysaccharides. Toxins (Basel) 2023; 15:394. [PMID: 37368695 DOI: 10.3390/toxins15060394] [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: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Using alternative feed ingredients in pig diets can lead to deoxynivalenol (DON) contamination. DON has been shown to induce anorexia, inflammation, and-more recently-alterations in the vitamin D, calcium, and phosphorus metabolisms. Adding vitamin D supplementation in the form of vitamin D3 and 25-OH-D3 to the feed could modify the effects of DON in piglets. In this study, vitamin D3 or 25-OH-D3 supplementation was used in a control or DON-contaminated treatment. A repetitive exposure over 21 days to DON in the piglets led to disruptions in the vitamin D, calcium, and phosphorus metabolisms, resulting in a decreased growth performance, increased bone mineralization, and the downregulation of genes related to calcium and to phosphorus intestinal and renal absorption. The DON challenge also decreased blood concentrations of 25-OH-D3, 1,25-(OH)2-D3, and phosphate. The DON contamination likely decreased the piglets' vitamin D status indirectly by modifying the calcium metabolism response. Vitamin D supplementations did not restore vitamin D status or bone mineralization. After a lipopolysaccharide-induced inflammatory stimulation, feeding a 25-OH-D3 supplementation increased 25-OH-D3 concentration and 1,25-(OH)2-D3 regulations during the DON challenge. DON contamination likely induced a Ca afflux by altering the intestinal barrier, which resulted in hypercalcemia and hypovitaminosis D. The vitamin D supplementation could increase the calcitriol production to face the combined LPS and DON challenge.
Collapse
Affiliation(s)
- Béatrice Sauvé
- Department of Animal Sciences, Laval University, Quebec, QC G1V 0A6, Canada
| | - Younes Chorfi
- Department of Veterinary Biomedicine, Montreal University, Saint-Hyacinthe, QC J2S 2M2, Canada
| | | | - Frédéric Guay
- Department of Animal Sciences, Laval University, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
40
|
Liang ZT, Li JK, Li J, Tang H, Guo CF, Zhang HQ. PECAM1 plays a role in the pathogenesis and treatment of bone metastases. Front Genet 2023; 14:1151651. [PMID: 37007939 PMCID: PMC10050551 DOI: 10.3389/fgene.2023.1151651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Bone is the third most common metastatic site for all primary tumors, the common primary focus of bone metastases include breast cancer, prostate cancer, and so on. And the median survival time of patients with bone metastases is only 2–3 years. Therefore, it is urgent to develop new targets to diagnose and treat bone metastases. Based on two data sets GSE146661 and GSE77930 associated with bone metastases, it was found that 209 genes differentially expressed in bone metastases group and control group. PECAM1 was selected as hub-gene for the follow-up research after constructing protein-protein interaction (PPI) network and enrichment analysis. Moreover, q-PCR analysis verified that the expression of PECAM1 decreased in bone metastatic tumor tissues. PECAM1 was believed to be possibly related to the function of osteoclasts, we knocked down the expression of PECAM1 with shRNA in lymphocytes extracted from bone marrow nailed blood. The results indicated that sh-PECAM1 treatment could promote osteoclast differentiation, and the sh-PECAM1-treated osteoclast culture medium could significantly promote the proliferation and migration of tumor cells. These results suggested that PECAM1 may be a potential biomarker for the diagnosis and treatment of bone metastases of tumor.
Collapse
Affiliation(s)
- Zhuo-Tao Liang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia-Ke Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jiong Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Tang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao-Feng Guo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Chao-Feng Guo, ; Hong-Qi Zhang,
| | - Hong-Qi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Chao-Feng Guo, ; Hong-Qi Zhang,
| |
Collapse
|
41
|
Weng Y, Jian Y, Huang W, Xie Z, Zhou Y, Pei X. Alkaline earth metals for osteogenic scaffolds: From mechanisms to applications. J Biomed Mater Res B Appl Biomater 2023; 111:1447-1474. [PMID: 36883838 DOI: 10.1002/jbm.b.35246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023]
Abstract
Regeneration of bone defects is a significant challenge today. As alternative approaches to the autologous bone, scaffold materials have remarkable features in treating bone defects; however, the various properties of current scaffold materials still fall short of expectations. Due to the osteogenic capability of alkaline earth metals, their application in scaffold materials has become an effective approach to improving their properties. Furthermore, numerous studies have shown that combining alkaline earth metals leads to better osteogenic properties than applying them alone. In this review, the physicochemical and physiological characteristics of alkaline earth metals are introduced, mainly focusing on their mechanisms and applications in osteogenesis, especially magnesium (Mg), calcium (Ca), strontium (Sr), and barium (Ba). Furthermore, this review highlights the possible cross-talk between pathways when alkaline earth metals are combined. Finally, some of the current drawbacks of scaffold materials are enumerated, such as the high corrosion rate of Mg scaffolds and defects in the mechanical properties of Ca scaffolds. Moreover, a brief perspective is also provided regarding future directions in this field. It is worth exploring that whether the levels of alkaline earth metals in newly regenerated bone differs from those in normal bone. The ideal ratio of each element in the bone tissue engineering scaffolds or the optimal concentration of each elemental ion in the created osteogenic environment still needs further exploration. The review not only summarizes the research developments in osteogenesis but also offers a direction for developing new scaffold materials.
Collapse
Affiliation(s)
- Yihang Weng
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Yujia Jian
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenlong Huang
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhuojun Xie
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Zhou
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Xibo Pei
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
42
|
Lungu O, Toscani D, Burroughs-Garcia J, Giuliani N. The Metabolic Features of Osteoblasts: Implications for Multiple Myeloma (MM) Bone Disease. Int J Mol Sci 2023; 24:ijms24054893. [PMID: 36902326 PMCID: PMC10003241 DOI: 10.3390/ijms24054893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The study of osteoblast (OB) metabolism has recently received increased attention due to the considerable amount of energy used during the bone remodeling process. In addition to glucose, the main nutrient for the osteoblast lineages, recent data highlight the importance of amino acid and fatty acid metabolism in providing the fuel necessary for the proper functioning of OBs. Among the amino acids, it has been reported that OBs are largely dependent on glutamine (Gln) for their differentiation and activity. In this review, we describe the main metabolic pathways governing OBs' fate and functions, both in physiological and pathological malignant conditions. In particular, we focus on multiple myeloma (MM) bone disease, which is characterized by a severe imbalance in OB differentiation due to the presence of malignant plasma cells into the bone microenvironment. Here, we describe the most important metabolic alterations involved in the inhibition of OB formation and activity in MM patients.
Collapse
Affiliation(s)
- Oxana Lungu
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Hematology, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy
- Correspondence:
| |
Collapse
|
43
|
Grčević D, Sanjay A, Lorenzo J. Interactions of B-lymphocytes and bone cells in health and disease. Bone 2023; 168:116296. [PMID: 34942359 PMCID: PMC9936888 DOI: 10.1016/j.bone.2021.116296] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/09/2023]
Abstract
Bone remodeling occurs through the interactions of three major cell lineages, osteoblasts, which mediate bone formation, osteocytes, which derive from osteoblasts, sense mechanical force and direct bone turnover, and osteoclasts, which mediate bone resorption. However, multiple additional cell types within the bone marrow, including macrophages, T lymphocytes and B lymphocytes influence the process. The bone marrow microenvironment, which is supported, in part, by bone cells, forms a nurturing network for B lymphopoiesis. In turn, developing B lymphocytes influence bone cells. Bone health during homeostasis depends on the normal interactions of bone cells with other lineages in the bone marrow. In disease state these interactions become pathologic and can cause abnormal function of bone cells and inadequate repair of bone after a fracture. This review summarizes what is known about the development of B lymphocytes and the interactions of B lymphocytes with bone cells in both health and disease.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, Croatian Institute for Brain Research, School of Medicine University of Zagreb, Zagreb, Croatia.
| | - Archana Sanjay
- Department of Orthopaedics, UConn Health, Farmington, CT, USA.
| | - Joseph Lorenzo
- Departments of Medicine and Orthopaedics, UConn Health, Farmington, CT, USA.
| |
Collapse
|
44
|
Teramachi J, Miki H, Nakamura S, Hiasa M, Harada T, Abe M. Myeloma bone disease: pathogenesis and management in the era of new anti-myeloma agents. J Bone Miner Metab 2023; 41:388-403. [PMID: 36856824 PMCID: PMC9975874 DOI: 10.1007/s00774-023-01403-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 03/02/2023]
Abstract
INTRODUCTION Multiple myeloma (MM) is a malignancy of plasma cells with characteristic bone disease. Despite recent great strides achieved in MM treatment owing to the implementation of new anti-MM agents, MM is still incurable and bone destruction remains a serious unmet issue in patients with MM. APPROACH In this review, we will summarize and discuss the mechanisms of the formation of bone disease in MM and the available preclinical and clinical evidence on the treatment for MM bone disease. CONCLUSIONS MM cells produce a variety of cytokines to stimulate receptor activator of nuclear factor-κB ligand-mediated osteoclastogenesis and suppress osteoblastic differentiation from bone marrow stromal cells, leading to extensive bone destruction with rapid loss of bone. MM cells alter the microenvironment through bone destruction where they colonize, which in turn favors tumor growth and survival, thereby forming a vicious cycle between tumor progression and bone destruction. Denosumab or zoledronic acid is currently recommended to be administered at the start of treatment in newly diagnosed patients with MM with bone disease. Proteasome inhibitors and the anti-CD38 monoclonal antibody daratumumab have been demonstrated to exert bone-modifying activity in responders. Besides their anti-tumor activity, the effects of new anti-MM agents on bone metabolism should be more precisely analyzed in patients with MM. Because prognosis in patients with MM has been significantly improved owing to the implementation of new agents, the therapeutic impact of bone-modifying agents should be re-estimated in the era of these new agents.
Collapse
Affiliation(s)
- Jumpei Teramachi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan.
- Department of Oral Function and Anatomy, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University Graduate School, 2-5-1 Shikata, Okayama, 700-8525, Japan.
| | - Hirokazu Miki
- Division of Transfusion Medicine and Cell Therapy, Tokushima University Hospital, Tokushima, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Hiasa
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan.
| |
Collapse
|
45
|
Peng H, Ling T, Zhang Y, Xie T, Pei X, Zhou K, Chen A, Chen J, Zhu X, Zhang X, Zhou Z. Nanowhiskers Orchestrate Bone Formation and Bone Defect Repair by Modulating Immune Cell Behavior. ACS APPLIED MATERIALS & INTERFACES 2023; 15:9120-9134. [PMID: 36755394 DOI: 10.1021/acsami.2c21865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immunomodulatory biomaterials have emerged as promising treatment agents for bone defects. However, it is unclear how such biomaterials control immune cell behaviors to facilitate large-segment bone defect repair. Herein, we fabricated biphasic calcium phosphate ceramics with nanowhisker structures to explore the immunoregulation features and influence on large-segment bone defect repair. We found that the nanowhisker structures markedly facilitated large-segment bone defect repair by promoting bone regeneration and scaffold resorption. Our in vitro experiment and transcriptomic analysis showed that mechanical stress derived from nanowhisker structures may activate the transcription of Egr-1 to induce early switch of macrophage phenotype to M2, which could not only facilitate osteogenic differentiation of BMSCs but also enhance the expression of osteoclast differentiation-regulating genes of M2 macrophage. In vivo study showed that the nanowhisker structures relieved local inflammatory responses by inducing early switch of macrophage phenotype from M1 to M2, which resulted in accelerated osteoclastogenesis for biomaterial resorption and osteogenesis for ectopic bone formation. Hence, we presume that nanowhisker structures may orchestrate bone formation and material resorption coupling to facilitate large-segment bone defect repair by controlling the switch of macrophage phenotype. This study provides new insight into the designing of immunomodulatory tissue engineering biomaterials for treating large-segment bone defects.
Collapse
Affiliation(s)
- Haitao Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tingxian Ling
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Zhang
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tianhang Xie
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuan Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Kai Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Anjing Chen
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zongke Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
46
|
Mumtaz N, Dudakovic A, Nair A, Koedam M, van Leeuwen JPTM, Koopmans MPG, Rockx B, van Wijnen AJ, van der Eerden BCJ. Zika virus alters osteogenic lineage progression of human mesenchymal stromal cells. J Cell Physiol 2023; 238:379-392. [PMID: 36538650 DOI: 10.1002/jcp.30933] [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: 02/03/2022] [Revised: 11/09/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
Arboviruses target bone forming osteoblasts and perturb bone remodeling via paracrine factors. We previously reported that Zika virus (ZIKV) infection of early-stage human mesenchymal stromal cells (MSCs) inhibited the osteogenic lineage commitment of MSCs. To understand the physiological interplay between bone development and ZIKV pathogenesis, we employed a primary in vitro model to examine the biological responses of MSCs to ZIKV infection at different stages of osteogenesis. Precommitted MSCs were infected at the late stage of osteogenic stimulation (Day 7) with ZIKV (multiplicity of infection of 5). We observe that MSCs infected at the late stage of differentiation are highly susceptible to ZIKV infection similar to previous observations with early stage infected MSCs (Day 0). However, in contrast to ZIKV infection at the early stage of differentiation, infection at a later stage significantly elevates the key osteogenic markers and calcium content. Comparative RNA sequencing (RNA-seq) of early and late stage infected MSCs reveals that ZIKV infection alters the mRNA transcriptome during osteogenic induction of MSCs (1251 genes). ZIKV infection provokes a robust antiviral response at both stages of osteogenic differentiation as reflected by the upregulation of interferon responsive genes (n > 140). ZIKV infection enhances the expression of immune-related genes in early stage MSCs while increasing cell cycle genes in late stage MSCs. Remarkably, ZIKA infection in early stage MSCs also activates lipid metabolism-related pathways. In conclusion, ZIKV infection has differentiation stage-dependent effects on MSCs and this mechanistic understanding may permit the development of new therapeutic or preventative measures for bone-related effects of ZIKV infection.
Collapse
Affiliation(s)
- Noreen Mumtaz
- Department of Viroscience, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Amel Dudakovic
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Asha Nair
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Johannes P T M van Leeuwen
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
47
|
Wang X, Shao L, Richardson KK, Ling W, Warren A, Krager K, Aykin-Burns N, Hromas R, Zhou D, Almeida M, Kim HN. Hematopoietic cytoplasmic adaptor protein Hem1 promotes osteoclast fusion and bone resorption in mice. J Biol Chem 2023; 299:102841. [PMID: 36574841 PMCID: PMC9867982 DOI: 10.1016/j.jbc.2022.102841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022] Open
Abstract
Hem1 (hematopoietic protein 1), a hematopoietic cell-specific member of the Hem family of cytoplasmic adaptor proteins, is essential for lymphopoiesis and innate immunity as well as for the transition of hematopoiesis from the fetal liver to the bone marrow. However, the role of Hem1 in bone cell differentiation and bone remodeling is unknown. Here, we show that deletion of Hem1 resulted in a markedly increase in bone mass because of defective bone resorption in mice of both sexes. Hem1-deficient osteoclast progenitors were able to differentiate into osteoclasts, but the osteoclasts exhibited impaired osteoclast fusion and decreased bone-resorption activity, potentially because of decreased mitogen-activated protein kinase and tyrosine kinase c-Abl activity. Transplantation of bone marrow hematopoietic stem and progenitor cells from wildtype into Hem1 knockout mice increased bone resorption and normalized bone mass. These findings indicate that Hem1 plays a pivotal role in the maintenance of normal bone mass.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lijian Shao
- Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kimberly K Richardson
- Division of Endocrinology, Department of Internal Medicine, Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Wen Ling
- Division of Endocrinology, Department of Internal Medicine, Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Aaron Warren
- Division of Endocrinology, Department of Internal Medicine, Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Robert Hromas
- Department of Medicine, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Daohong Zhou
- Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Maria Almeida
- Division of Endocrinology, Department of Internal Medicine, Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Ha-Neui Kim
- Division of Endocrinology, Department of Internal Medicine, Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
48
|
Welsh H, Brickley MB. Pathology or expected morphology? Investigating patterns of cortical porosity and trabecularization during infancy and early childhood. Anat Rec (Hoboken) 2023; 306:354-365. [PMID: 36116138 DOI: 10.1002/ar.25081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/15/2022] [Accepted: 09/08/2022] [Indexed: 01/25/2023]
Abstract
Increased cortical porosity is associated with a heightened risk of skeletal fragility due to bone loss and structural decay in adults. However, few studies have examined the etiology of cortical porosity in infants and children. This study examines whether age-related changes in femoral growth and locomotor development influence femoral midshaft cortical porosity in a sample of 48 individuals (fetal to 3.99 years) from the 10th-13th century cemetery of St. Étienne de Toulouse, France. Histological sections were prepared and imaged using light microscopy. Midshaft geometric variables such as total area, cortical area, and pore area were calculated using BoneJ. Increased porosity and cortical trabecularization were found to be significantly associated with age, being almost exclusively present in individuals aged 0.5-1.99 years. At approximately 6 months of age infants typically begin engaging in regular femoral loading and experience an acceleration in growth. The observed increase in midshaft porosity and trabecularization, therefore, likely results from the reorganization and redistribution of cortical bone, stimulated by increased growth velocity and the onset of weight-bearing activities. The reduction in cortical porosity and trabecularization in individuals aged 2.0-3.99 years indicates that children are approaching some sort of homeostasis as growth velocity slows and their femora adapt to consistent loading. Understanding what expected skeletal development looks like is necessary when conducting bioarcheological studies and this study provides evidence for a pattern of transient midshaft porosity during infancy and early childhood.
Collapse
Affiliation(s)
- Hayley Welsh
- Department of Anthropology, University of Toronto, Toronto, Ontario, Canada
| | - Megan B Brickley
- Department of Anthropology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
49
|
Albrektsson T, Tengvall P, Amengual L, Coli P, Kotsakis GA, Cochran D. Osteoimmune regulation underlies oral implant osseointegration and its perturbation. Front Immunol 2023; 13:1056914. [PMID: 36761175 PMCID: PMC9902598 DOI: 10.3389/fimmu.2022.1056914] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/20/2022] [Indexed: 01/26/2023] Open
Abstract
In the field of biomaterials, an endosseous implant is now recognized as an osteoimmunomodulatory but not bioinert biomaterial. Scientific advances in bone cell biology and in immunology have revealed a close relationship between the bone and immune systems resulting in a field of science called osteoimmunology. These discoveries have allowed for a novel interpretation of osseointegration as representing an osteoimmune reaction rather than a classic bone healing response, in which the activation state of macrophages ((M1-M2 polarization) appears to play a critical role. Through this viewpoint, the immune system is responsible for isolating the implant biomaterial foreign body by forming bone around the oral implant effectively shielding off the implant from the host bone system, i.e. osseointegration becomes a continuous and dynamic host defense reaction. At the same time, this has led to the proposal of a new model of osseointegration, the foreign body equilibrium (FBE). In addition, as an oral wound, the soft tissues are involved with all their innate immune characteristics. When implant integration is viewed as an osteoimmune reaction, this has implications for how marginal bone is regulated. For example, while bacteria are constitutive components of the soft tissue sulcus, if the inflammatory front and immune reaction is at some distance from the marginal bone, an equilibrium is established. If however, this inflammation approaches the marginal bone, an immune osteoclastic reaction occurs and marginal bone is removed. A number of clinical scenarios can be envisioned whereby the osteoimmune equilibrium is disturbed and marginal bone loss occurs, such as complications of aseptic nature and the synergistic activation of pro-inflammatory pathways (implant/wear debris, DAMPs, and PAMPs). Understanding that an implant is a foreign body and that the host reacts osteoimmunologically to shield off the implant allows for a distinction to be drawn between osteoimmunological conditions and peri-implant bone loss. This review will examine dental implant placement as an osteoimmune reaction and its implications for marginal bone loss.
Collapse
Affiliation(s)
- T. Albrektsson
- Department of Biomaterials, University of Gothenburg, Gothenburg, Sweden
| | - P. Tengvall
- Department of Biomaterials, University of Gothenburg, Gothenburg, Sweden,*Correspondence: P. Tengvall,
| | - L. Amengual
- Dental Implantology Unit, Hospital Leonardo Guzmán, Antofagasta, Chile
| | - P. Coli
- Edinburgh Dental Specialists, Edinburgh, United Kingdom,Department of Prosthetic Dentistry and Dental Material Science, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden,Department of Dental Material Science, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - G. A. Kotsakis
- Department of Periodontology, University of Texas, San Antonio, TX, United States
| | - D. Cochran
- Department of Periodontology, University of Texas, San Antonio, TX, United States
| |
Collapse
|
50
|
Calvo-Gallego JL, Manchado-Morales P, Pivonka P, Martínez-Reina J. Spatio-temporal simulations of bone remodelling using a bone cell population model based on cell availability. Front Bioeng Biotechnol 2023; 11:1060158. [PMID: 36959906 PMCID: PMC10027742 DOI: 10.3389/fbioe.2023.1060158] [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: 10/02/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Here we developed a spatio-temporal bone remodeling model to simulate the action of Basic Multicelluar Units (BMUs). This model is based on two major extensions of a temporal-only bone cell population model (BCPM). First, the differentiation into mature resorbing osteoclasts and mature forming osteoblasts from their respective precursor cells was modelled as an intermittent process based on precursor cells availability. Second, the interaction between neighbouring BMUs was considered based on a "metabolic cost" argument which warrants that no new BMU will be activated in the neighbourhood of an existing BMU. With the proposed model we have simulated the phases of the remodelling process obtaining average periods similar to those found in the literature: resorption ( ∼ 22 days)-reversal (∼8 days)-formation (∼65 days)-quiescence (560-600 days) and an average BMU activation frequency of ∼1.6 BMUs/year/mm3. We further show here that the resorption and formation phases of the BMU become coordinated only by the presence of TGF-β (transforming growth factor β), i.e., a major coupling factor stored in the bone matrix. TGF-β is released through resorption so upregulating osteoclast apoptosis and accumulation of osteoblast precursors, i.e., facilitating the transition from the resorption to the formation phase at a given remodelling site. Finally, we demonstrate that this model can explain targeted bone remodelling as the BMUs are steered towards damaged bone areas in order to commence bone matrix repair.
Collapse
Affiliation(s)
- José Luis Calvo-Gallego
- Departamento de Ingeniería Mecánica y Fabricación, Universidad de Sevilla, Seville, Spain
- *Correspondence: José Luis Calvo-Gallego,
| | - Pablo Manchado-Morales
- Departamento de Ingeniería Mecánica y Fabricación, Universidad de Sevilla, Seville, Spain
| | - Peter Pivonka
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Javier Martínez-Reina
- Departamento de Ingeniería Mecánica y Fabricación, Universidad de Sevilla, Seville, Spain
| |
Collapse
|