1
|
Ivanovic-Zuvic D, Madison A, Jiménez M, Oyanadel ML, Gattini N, Guzmán C, Cifré M, Silvestre R, García C, Contreras O, Collins MT, Florenzano P. Factors associated with impaired physical functionality in X-linked hypophosphatemia. JBMR Plus 2025; 9:ziaf018. [PMID: 40092459 PMCID: PMC11910016 DOI: 10.1093/jbmrpl/ziaf018] [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: 08/19/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
X-linked hypophosphatemia (XLH) is a rare inherited disorder characterized by elevated levels of FGF23, chronic hypophosphatemia, impaired bone mineralization, and chronic long-term manifestations. Treatment for XLH has been mainly focused on normalizing its biochemical abnormalities. Despite treatment, patients with XLH often present impaired physical function and decreased quality of life (QoL). We hypothesize that physical functionality and QoL are more strongly associated with chronic pain and decreased muscle mass than persistent biochemical abnormalities or exposure to conventional treatment. We conducted an observational, cross-sectional study with patients with XLH. Clinical records and biochemical parameters were assessed. QoL surveys SF36v.2 and WOMAC were applied. Functional status was measured by a physiatrist and an occupational therapist. Appendicular lean mass (ALM) was measured and compared with age and sex-matched healthy controls. Enthesopathies and osteoarthritis were evaluated. Pain was assessed using the Brief Pain Inventory, the Visual Analog Scale, and the Doleur Neuropathique-4 scales. Muscle strength was evaluated by the quadriceps muscle isometric strength (QMS) and physical performance with the 6-Minute Walk Test (6MWT) and the Functional Independence Measure (FIM) scale. A total of 30 patients were included: 21 females; median age of 32 yr. All participants had significant functional deficits, chronic pain, and reduced QoL. Limitations in daily activities were significantly associated with higher severity of pain, decreased ALM, lower QMS, and less distance in 6MWT (p < .05). Neither FIM scale, phosphate levels, FGF23, nor the lifetime exposure to conventional treatment was associated with these functional variables. In conclusion, impaired physical functionality in patients with XLH was associated with lower muscle mass, lower muscle strength, and severe chronic pain. These findings highlight the importance of, in addition to optimizing the biochemical control of the disease, expanding patient care including pain prevention and management as well as comprehensive physical therapy and rehabilitation.
Collapse
Affiliation(s)
| | - Annette Madison
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| | - Macarena Jiménez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| | - María L Oyanadel
- Department of Physical Medicine and Rehabilitation, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
- Department of Physical Medicine and Rehabilitation, San Borja Arriarán Hospital, Santiago, 8360160, Chile
| | - Natalia Gattini
- Department of Physical Medicine and Rehabilitation, Hospital Clínico Universidad de Chile, Santiago, 8380000, Chile
| | - Carolina Guzmán
- Department of Physical Medicine and Rehabilitation, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| | - Manuela Cifré
- Biomechanics Unit, Innovation Center, Clínica MEDS, Santiago, 7550557, Chile
| | - Rony Silvestre
- Biomechanics Unit, Innovation Center, Clínica MEDS, Santiago, 7550557, Chile
- Musculoskeletal Department, Kinesiology School, Pontificia Universidad Católica, Santiago, 7820609, Chile
| | - Cristián García
- Department of Radiology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| | - Oscar Contreras
- Department of Radiology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| | - Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Pablo Florenzano
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
- Skeletal Disorders and Mineral Homeostasis Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD 20892, United States
- Department of Endocrinology, Center for Translational Research in Endocrinology, CETREN-UC, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330023, Chile
| |
Collapse
|
2
|
Spoladore R, Ciampi CM, Ossola P, Sultana A, Spreafico LP, Farina A, Fragasso G. Heart Failure and Osteoporosis: Shared Challenges in the Aging Population. J Cardiovasc Dev Dis 2025; 12:69. [PMID: 39997503 PMCID: PMC11856909 DOI: 10.3390/jcdd12020069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
In clinical practice, heart failure (HF) and osteoporosis (OP) are commonly paired conditions. This association is particularly relevant in patients over the age of 50, among whom its prevalence increases dramatically with every decade of life. This can be especially impactful since patient prognosis when facing both conditions is poorer than that of each disease alone. Clinical studies suggest that prior fractures increase the risk for heart failure hospitalization and, conversely, an episode of heart failure increases the risk of subsequent fractures. In other words, the relationship between osteoporosis and heart failure seems to be two-way, meaning that each condition may influence or contribute to the development of the other. However, the details of the pathophysiological relationship between HF and OP have yet to be revealed. The two conditions share multiple pathological mechanisms that seem to be intertwined. Patients affected by OP are more prone to develop HF because of vitamin D deficiency, elevation of parathyroid hormone (PTH) plasma levels, and increased Fibroblast Growth Factor 23 (FGF-23) activity. On the other hand, HF patients are more prone to develop OP and pathological fractures because of low vitamin D level, high PTH, chronic renal failure, alteration of renin-angiotensin-aldosterone system, reduced testosterone level, and metabolic effects derived from commonly used medications. Considering the increasingly aging worldwide population, clinicians can expect to see more often an overlap between these two conditions. Thus, it becomes crucial to recognize how HF and OP mutually influence the patient's clinical condition. Clinicians attending these patients should utilize an integrated approach and, in order to improve prognosis, aim for early diagnosis and treatment initiation. The aim of this paper is to perform a review of the common pathophysiological mechanisms of OP and HF and identify potentially new treatment targets.
Collapse
Affiliation(s)
- Roberto Spoladore
- Heart Failure Clinic, Division of Cardiology, Alessandro Manzoni Hospital, ASST Lecco, 23900 Lecco, Italy;
| | - Claudio Mario Ciampi
- Health Science Department, University of Milan Bicocca, 20126 Milan, Italy; (C.M.C.); (P.O.); (A.S.)
| | - Paolo Ossola
- Health Science Department, University of Milan Bicocca, 20126 Milan, Italy; (C.M.C.); (P.O.); (A.S.)
| | - Andrea Sultana
- Health Science Department, University of Milan Bicocca, 20126 Milan, Italy; (C.M.C.); (P.O.); (A.S.)
| | - Luigi Paolo Spreafico
- Orthopedics and Traumatology Unit, San Paolo University Hospital, 20142 Milan, Italy;
| | - Andrea Farina
- Heart Failure Clinic, Division of Cardiology, Alessandro Manzoni Hospital, ASST Lecco, 23900 Lecco, Italy;
| | - Gabriele Fragasso
- Heart Failure Clinic, Division of Cardiology, IRCCS San Raffaele University Hospital, 20132 Milan, Italy;
| |
Collapse
|
3
|
Lin W, Li Y, Qiu C, Zou B, Gong Y, Zhang X, Tian D, Sherman W, Sanchez F, Wu D, Su KJ, Xiao X, Luo Z, Tian Q, Chen Y, Shen H, Deng H. Mapping the spatial atlas of the human bone tissue integrating spatial and single-cell transcriptomics. Nucleic Acids Res 2025; 53:gkae1298. [PMID: 39817519 PMCID: PMC11736439 DOI: 10.1093/nar/gkae1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Bone is a multifaceted tissue requiring orchestrated interplays of diverse cells within specialized microenvironments. Although significant progress has been made in understanding cellular and molecular mechanisms of component cells of bone, revealing their spatial organization and interactions in native bone tissue microenvironment is crucial for advancing precision medicine, as they govern fundamental signaling pathways and functional dependencies among various bone cells. In this study, we present the first integrative high-resolution map of human bone and bone marrow, using spatial and single-cell transcriptomics profiling from femoral tissue. This multi-modal approach discovered a novel bone formation-specialized niche enriched with osteoblastic lineage cells and fibroblasts and unveiled critical cell-cell communications and co-localization patterns between osteoblastic lineage cells and other cells. Furthermore, we discovered a novel spatial gradient of cellular composition, gene expression and signaling pathway activities radiating from the trabecular bone. This comprehensive atlas delineates the intricate bone cellular architecture and illuminates key molecular processes and dependencies among cells that coordinate bone metabolism. In sum, our study provides an essential reference for the field of bone biology and lays the foundation for advanced mechanistic studies and precision medicine approaches in bone-related disorders.
Collapse
Affiliation(s)
- Weiqiang Lin
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yisu Li
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Binghao Zou
- Department of Structural and Cellular Biology, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xiao Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Di Tian
- The Molecular Pathology Laboratory, Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - William Sherman
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Fernando Sanchez
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Di Wu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xinyi Xiao
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Zhe Luo
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yiping Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Hongwen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| |
Collapse
|
4
|
Rummler M, van Tol A, Schemenz V, Hartmann MA, Blouin S, Willie BM, Weinkamer R. The Lacunocanalicular Network is Denser in C57BL/6 Compared to BALB/c Mice. Calcif Tissue Int 2024; 115:744-758. [PMID: 39414712 PMCID: PMC11531440 DOI: 10.1007/s00223-024-01289-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: 02/07/2024] [Accepted: 09/10/2024] [Indexed: 10/18/2024]
Abstract
The lacunocanalicular network (LCN) is an intricate arrangement of cavities (lacunae) and channels (canaliculi), which permeates the mineralized bone matrix. In its porosity, the LCN accommodates the cell network of osteocytes. These two nested networks are attributed a variety of essential functions including transport, signaling, and mechanosensitivity due to load-induced fluid flow through the LCN. For a more quantitative assessment of the networks' function, the three-dimensional architecture has to be known. For this reason, we aimed (i) to quantitatively characterize spatial heterogeneities of the LCN in whole mouse tibial cross-sections of BALB/c mice and (ii) to analyze differences in LCN architecture by comparison with another commonly used inbred mouse strain, the C57BL/6 mouse. Both tibiae of five BALB/c mice (female, 26-week-old) were stained using rhodamine 6G and whole tibiae cross-sections were imaged using confocal laser scanning microscopy. Using image analysis, the LCN was quantified in terms of density and connectivity and lacunar parameters, such as lacunar degree, volume, and shape. In the same tibial cross-sections, the calcium content was measured using quantitative backscattered electron imaging (qBEI). A structural analysis of the LCN properties showed that spatially denser parts of the LCN are mainly due to a higher density of branching points in the network. While a high intra-individual variability of network density was detected within the cortex, the inter-individual variability between different mice was low. In comparison to C57BL/6J mice, BALB/c mice showed a distinct lower canalicular density. This reduced network was already detectable on a local network level with fewer canaliculi emanating from lacunae. Spatial correlation with qBEI images demonstrated that bone modeling resulted in disruptions in the network architecture. The spatial heterogeneity and differences in density of the LCN likely affects the fluid flow within the network and therefore bone's mechanoresponse to loading.
Collapse
Affiliation(s)
- Maximilian Rummler
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
| | - Alexander van Tol
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
| | - Victoria Schemenz
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany
- Department of Operative and Preventive Dentistry, Charité-Universitätsmedizin - Berlin, Berlin, Germany
| | - Markus A Hartmann
- 1st Medical Department Hanusch Hospital, Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Vienna, Austria
| | - Stéphane Blouin
- 1st Medical Department Hanusch Hospital, Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Vienna, Austria
| | - Bettina M Willie
- Research Center, Shriners Hospital for Children, Montreal, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Richard Weinkamer
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany.
| |
Collapse
|
5
|
Yin Y, Chen G, Yang C, Wang J, Peng J, Huang X, Tang Q, Chen L. Osteocyte ferroptosis induced by ATF3/TFR1 contributes to cortical bone loss during ageing. Cell Prolif 2024; 57:e13657. [PMID: 38764128 PMCID: PMC11471391 DOI: 10.1111/cpr.13657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Cortical bone loss is intricately associated with ageing and coincides with iron accumulation. The precise role of ferroptosis, characterized by iron overload and lipid peroxidation, in senescent osteocytes remains elusive. We found that ferroptosis was a crucial mode of osteocyte death in cortical bone during ageing. Using a single-cell transcriptome analysis, we identified activating transcription factor 3 (ATF3) as a critical driver of osteocyte ferroptosis. Elevated ATF3 expression in senescent osteocytes promotes iron uptake by upregulating transferrin receptor 1 while simultaneously inhibiting solute carrier family 7-member 11-mediated cystine import. This process leads to an iron overload and lipid peroxidation, culminating in ferroptosis. Importantly, ATF3 inhibition in aged mice effectively alleviated ferroptosis in the cortical bone and mitigated cortical bone mass loss. Taken together, our findings establish a pivotal role of ferroptosis in cortical bone loss in older adults, providing promising prevention and treatment strategies for osteoporosis and fractures.
Collapse
Affiliation(s)
- Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Guang‐Jin Chen
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Chen Yang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Jia‐Jia Wang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Jin‐Feng Peng
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Xiao‐Fei Huang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Qing‐Ming Tang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Li‐Li Chen
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
6
|
Chen PJ, Mehta S, Dutra EH, Yadav S. Alendronate treatment rescues the effects of compressive loading of TMJ in osteogenesis imperfecta mice. Prog Orthod 2024; 25:25. [PMID: 39004686 PMCID: PMC11247069 DOI: 10.1186/s40510-024-00526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is a genetic disorder of connective tissue caused by mutations associated with type I collagen, which results in defective extracellular matrix in temporomandibular joint (TMJ) cartilage and subchondral bone. TMJ is a fibrocartilaginous joint expressing type I collagen both in the cartilage and the subchondral bone. In the present study the effects of alendronate and altered loading of the TMJ was analyzed both in male and female OI mice. MATERIALS AND METHODS Forty-eight, 10-weeks-old male and female OI mice were divided into 3 groups: (1) Control group: unloaded group, (2) Saline + Loaded: Saline was injected for 2 weeks and then TMJ of mice was loaded for 5 days, (3) alendronate + loaded: alendronate was injected for 2 weeks and then TMJ of mice was loaded for 5 days. Mice in all the groups were euthanized 24-h after the final loading. RESULTS Alendronate pretreatment led to significant increase in bone volume and tissue density. Histomorphometrically, alendronate treatment led to increase in mineralization, cartilage thickness and proteoglycan distribution. Increased mineralization paralleled decreased osteoclastic activity. Our immunohistochemistry revealed decreased expression of matrix metallopeptidase 13 and ADAM metallopeptidase with thrombospondin type 1 motif 5. CONCLUSION The findings of this research support that alendronate prevented the detrimental effects of loading on the extracellular matrix of the TMJ cartilage and subchondral bone.
Collapse
Affiliation(s)
- Po-Jung Chen
- Department of Growth and Development, College of Dentistry, University of Nebraska Medical Center, Lincoln/Omaha, NE, USA.
| | - Shivam Mehta
- Department of Orthodontics, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Eliane H Dutra
- Division of Orthodontics, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Sumit Yadav
- Department of Growth and Development, College of Dentistry, University of Nebraska Medical Center, Lincoln/Omaha, NE, USA
| |
Collapse
|
7
|
Li X, Zhao J, Chen L, Zhou X, Qiu M, Deng L, Yang K, Xu Y. HIF-1α activation impairs dendrites formation and elongation in osteocytogenesis. Heliyon 2024; 10:e32889. [PMID: 39005918 PMCID: PMC11239586 DOI: 10.1016/j.heliyon.2024.e32889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Osteocytes are terminally differentiated cells derived from osteoblasts and are deeply embedded within the bone matrix. They play a critical role in bone remodeling by generating a lacuno-canalicular network (LCN) and controlling the transport of nutrients. Due to the absence of blood vessels within the bone matrix, it is widely believed that osteocytes develop in a hypoxic environment. However, the mechanisms of osteocytogenesis and the role of oxygen sensing in this process remain unclear. Hypoxia-inducible factors (HIFs) are major transcriptional factors involved in oxygen sensing. Previous studies have shown that accumulation of HIFs in osteoblasts leads to abnormal bone remodeling, potentially linked with the alterations of the LCN network. Specifically, HIF-1α is hypothesized to play a more significant role in regulating bone remodeling compared to HIF-2α. Therefore, we investigated the functions of HIF-1α in dendrite formation and the establishment of the LCN network during osteocytogenesis. Immunostaining and scanning electron microscopy revealed that the E11 protein aggregates to form a ring structure that defines the site for dendrite initiation. This process is followed by activation of the ERM/RhoA pathway and recruitment of matrix metalloproteinase 14 (MMP14) to facilitate extracellular matrix degradation, enabling dendrite elongation. However, both hypoxic treatment and overexpression of HIF-1α impair ring formation, resulting in reduced ERM/RhoA activity and decreased matrix degradation capability. These findings suggest that abnormal HIF-1α activity in local areas could contribute to impaired LCN network formation and abnormal bone remodeling observed in bone diseases such as osteopenia and aging.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272029, Shandong Province, China
| | - Jian Zhao
- Department of Orthopaedics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Lei Chen
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272029, Shandong Province, China
| | - Xinyi Zhou
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Minglong Qiu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Kai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
8
|
Jiang Z, Qi G, He X, Yu Y, Cao Y, Zhang C, Zou W, Yuan H. Ferroptosis in Osteocytes as a Target for Protection Against Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307388. [PMID: 38233202 PMCID: PMC10966575 DOI: 10.1002/advs.202307388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Ferroptosis is a necrotic form of iron-dependent regulatory cell death. Estrogen withdrawal can interfere with iron metabolism, which is responsible for the pathogenesis of postmenopausal osteoporosis (PMOP). Here, it is demonstrated that estrogen withdrawal induces iron accumulation in the skeleton and the ferroptosis of osteocytes, leading to reduced bone mineral density. Furthermore, the facilitatory effect of ferroptosis of osteocytes is verified in the occurrence and development of postmenopausal osteoporosis is associated with over activated osteoclastogenesis using a direct osteocyte/osteoclast coculture system and glutathione peroxidase 4 (GPX4) knockout ovariectomized mice. In addition, the nuclear factor erythroid derived 2-related factor-2 (Nrf2) signaling pathway is confirmed to be a crucial factor in the ferroptosis of osteocytic cells. Nrf2 regulates the expression of nuclear factor kappa-B ligand (RANKL) by regulating the DNA methylation level of the RANKL promoter mediated by DNA methyltransferase 3a (Dnmt3a), which is as an important mechanism in osteocytic ferroptosis-mediated osteoclastogenesis. Taken together, this data suggests that osteocytic ferroptosis is involved in PMOP and can be targeted to tune bone homeostasis.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Guobin Qi
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xuecheng He
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yifan Yu
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuting Cao
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Changqing Zhang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Weiguo Zou
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell SciencesShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Hengfeng Yuan
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
9
|
Al-Suhaimi EA, Akhtar S, Al Hubail FA, Alhawaj H, Aljafary MA, Alrumaih HS, Daghestani A, Al-Buainain A, Lardhi A, Homeida AM. A crosstalk between 'osteocyte lacunal-canalicular system' and metabolism. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 142:397-420. [PMID: 39059992 DOI: 10.1016/bs.apcsb.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Considering the importance, bone physiology has long been studied to understand what systematic and cellular impact its cells and functions have. Exploring more questions is a substantially solid way to improve the understanding of bone physiological functions in/out sides. In adult bone, osteocytes (Ots) form about 95% of bone cells and live the longest lifespan inside their mineralized surroundings. Ots are the endocrine cells and originate from blood vessel's endothelial cells. In this work, we discussed the vital role of the "Ots". To determine the association between osteocytes' network with metabolic parameters in healthy mice, the experiments were performed on ten (10) adult C57BL6 male mice. Fasting blood and bone samples were collected weekly from mice for measurement of metabolic parameters and bone morphology. Scanning electron microscopy (SEM) revealed a 2D fine morphology of the bone which indicates a strong functional interconnection with bone nano/micro, and macro components of the organs. The long-branched canaliculi look like neurocytes in structure. The morphology and quantitative measurements of the osteocyte lacunal-canalicular system showed its wide spectrum spatial resolution of the positive and negative relationship within this system or metabolite parameters, confirming a strong cross connection between osteocyte lacunal-canalicular system and metabolism. We believe that the findings of this study can deliver a strategy about the potential roles of metabolic relation among osteocytes, insulin, and lipid in management of bone and metabolic diseases.
Collapse
Affiliation(s)
- Ebtesam A Al-Suhaimi
- Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia; Department of Environmental Health Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Sultan Akhtar
- Department of Biophysics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Fatima A Al Hubail
- Mawhiba Research Enrichment Program-2022, King Abdulaziz and His Companions Foundation for Giftedness and Creativity, Riyadh, Saudi Arabia
| | - Hussain Alhawaj
- Department of Environmental Health Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Meneerah A Aljafary
- Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hamad S Alrumaih
- Department of Substitutive Dental Science, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Amira Daghestani
- Department of Substitutive Dental Science, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Alanwood Al-Buainain
- College of Science and Human Studies in Jubail, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Amer Lardhi
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - A M Homeida
- Department of Environmental Health Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
10
|
Lv Z, Zhang J, Liang S, Zhou C, Hu D, Brooks DJ, Bouxsein ML, Lanske B, Kostenuik P, Gori F, Baron R. Comparative study in estrogen-depleted mice identifies skeletal and osteocyte transcriptomic responses to abaloparatide and teriparatide. JCI Insight 2023; 8:e161932. [PMID: 37870958 PMCID: PMC10619488 DOI: 10.1172/jci.insight.161932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/08/2023] [Indexed: 10/25/2023] Open
Abstract
Osteocytes express parathyroid hormone (PTH)/PTH-related protein (PTHrP) receptors and respond to the PTHrP analog abaloparatide (ABL) and to the PTH 1-34 fragment teriparatide (TPTD), which are used to treat osteoporosis. Several studies indicate overlapping but distinct skeletal responses to ABL or TPTD, but their effects on cortical bone may differ. Little is known about their differential effects on osteocytes. We compared cortical osteocyte and skeletal responses to ABL and TPTD in sham-operated and ovariectomized mice. Administered 7 weeks after ovariectomy for 4 weeks at a dose of 40 μg/kg/d, TPTD and ABL had similar effects on trabecular bone, but ABL showed stronger effects in cortical bone. In cortical osteocytes, both treatments decreased lacunar area, reflecting altered peri-lacunar remodeling favoring matrix accumulation. Osteocyte RNA-Seq revealed that several genes and pathways were altered by ovariectomy and affected similarly by TPTD and ABL. Notwithstanding, several signaling pathways were uniquely regulated by ABL. Thus, in mice, TPTD and ABL induced a positive osteocyte peri-lacunar remodeling balance, but ABL induced stronger cortical responses and affected the osteocyte transcriptome differently. We concluded that ABL affected the cortical osteocyte transcriptome in a manner subtly different from TPTD, resulting in more beneficial remodeling/modeling changes and homeostasis of the cortex.
Collapse
Affiliation(s)
- Zhengtao Lv
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Jiaming Zhang
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Shuang Liang
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Chenhe Zhou
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Dorothy Hu
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Daniel J. Brooks
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Mary L. Bouxsein
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School and Massachusetts General Hospital (MGH) Endocrine Unit, Boston, Massachusetts, USA
| | | | | | - Francesca Gori
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Roland Baron
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
- Harvard Medical School and Massachusetts General Hospital (MGH) Endocrine Unit, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Laster M, Pereira RC, Noche K, Gales B, Salusky IB, Albrecht LV. Sclerostin, Osteocytes, and Wnt Signaling in Pediatric Renal Osteodystrophy. Nutrients 2023; 15:4127. [PMID: 37836411 PMCID: PMC10574198 DOI: 10.3390/nu15194127] [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: 08/05/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
The pathophysiology of chronic kidney disease-mineral and bone disorder (CKD-MBD) is not well understood. Specific factors secreted by osteocytes are elevated in the serum of adults and pediatric patients with CKD-MBD, including FGF-23 and sclerostin, a known inhibitor of the Wnt signaling pathway. The molecular mechanisms that promote bone disease during the progression of CKD are incompletely understood. In this study, we performed a cross-sectional analysis of 87 pediatric patients with pre-dialysis CKD and post-dialysis (CKD 5D). We assessed the associations between serum and bone sclerostin levels and biomarkers of bone turnover and bone histomorphometry. We report that serum sclerostin levels were elevated in both early and late CKD. Higher circulating and bone sclerostin levels were associated with histomorphometric parameters of bone turnover and mineralization. Immunofluorescence analyses of bone biopsies evaluated osteocyte staining of antibodies towards the canonical Wnt target, β-catenin, in the phosphorylated (inhibited) or unphosphorylated (active) forms. Bone sclerostin was found to be colocalized with phosphorylated β-catenin, which suggests that Wnt signaling was inhibited. In patients with low serum sclerostin levels, increased unphosphorylated "active" β-catenin staining was observed in osteocytes. These data provide new mechanistic insight into the pathogenesis of CKD-MBD and suggest that sclerostin may offer a potential biomarker or therapeutic target in pediatric renal osteodystrophy.
Collapse
Affiliation(s)
- Marciana Laster
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA; (M.L.); (R.C.P.); (K.N.); (B.G.)
| | - Renata C. Pereira
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA; (M.L.); (R.C.P.); (K.N.); (B.G.)
| | - Kathleen Noche
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA; (M.L.); (R.C.P.); (K.N.); (B.G.)
| | - Barbara Gales
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA; (M.L.); (R.C.P.); (K.N.); (B.G.)
| | - Isidro B. Salusky
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA; (M.L.); (R.C.P.); (K.N.); (B.G.)
| | - Lauren V. Albrecht
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, CA 92697, USA
| |
Collapse
|
12
|
Zhou YH, Zhu JY, Guo Y, Tang HN, Wang F, Iqbal J, Wu HX, Hu N, Xiao F, Wang T, Li L, Zhou HD. Notch1 is a marker for in situ resting osteocytes in a 3-dimensional gel culture model. Connect Tissue Res 2023; 64:491-504. [PMID: 37227119 DOI: 10.1080/03008207.2023.2217271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
PURPOSE Osteocytes in vivo exhibit different functional states, but no specific marker to distinguish these is currently available. MATERIALS AND METHODS To simulate the differentiation process of pre-osteoblasts to osteocytes in vitro, MC3T3-E1 cells were cultured on type I collagen gel and a three-dimensional (3D) culture system was established. The Notch expression of osteocyte-like cells in 3D culture system was compared with that of in situ osteocytes in bone tissues. RESULTS Immunohistochemistry demonstrated that Notch1 was not detected in "resting" in situ osteocytes, but was detected in normal cultured osteocyte-like cell line MLO-Y4. Osteocytes obtained from conventional osteogenic-induced osteoblasts and long-term cultured MLO-Y4 cells could not replicate the Notch1 expression pattern from in situ osteocytes. From day 14-35 of osteogenic induction, osteoblasts in 3D culture system gradually migrated into the gel to form canaliculus-like structures similar to bone canaliculus. On day 35, stellate-shaped osteocyte-like cells were observed, and expression of DMP1 and SOST, but not Runx2, was detected. Notch1 was not detected by immunohistochemistry, and Notch1 mRNA level was not significantly different from that of in situ osteocytes. In MC3T3-E1 cells, down-regulation of Notch2 increased Notch1, Notch downstream genes (β-catenin and Nfatc1), and Dmp1. In MLO-Y4 cells, Notch2 decreased after Notch1 siRNA transfection. Downregulation of Notch1 or Notch2 decreased Nfatc1, β-catenin, and Dmp1, and increased Sost. CONCLUSIONS We established "resting state" osteocytes using an in vitro 3D model. Notch1 can be a useful marker to help differentiate the functional states of osteocytes (activated vs. resting state).
Collapse
Affiliation(s)
- Ying-Hui Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Stomatology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jia-Yu Zhu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yue Guo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Stomatology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hao-Neng Tang
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fang Wang
- Departments of Endocrinology and Metabolism, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Junaid Iqbal
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Nan Hu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ting Wang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Lipreri MV, Di Pompo G, Boanini E, Graziani G, Sassoni E, Baldini N, Avnet S. Bone on-a-chip: a 3D dendritic network in a screening platform for osteocyte-targeted drugs. Biofabrication 2023; 15:045019. [PMID: 37552982 DOI: 10.1088/1758-5090/acee23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
Age-related musculoskeletal disorders, including osteoporosis, are frequent and associated with long lasting morbidity, in turn significantly impacting on healthcare system sustainability. There is therefore a compelling need to develop reliable preclinical models of disease and drug screening to validate novel drugs possibly on a personalized basis, without the need ofin vivoassay. In the context of bone tissue, although the osteocyte (Oc) network is a well-recognized therapeutic target, currentin vitropreclinical models are unable to mimic its physiologically relevant and highly complex structure. To this purpose, several features are needed, including an osteomimetic extracellular matrix, dynamic perfusion, and mechanical cues (e.g. shear stress) combined with a three-dimensional (3D) culture of Oc. Here we describe, for the first time, a high throughput microfluidic platform based on 96-miniaturized chips for large-scale preclinical evaluation to predict drug efficacy. We bioengineered a commercial microfluidic device that allows real-time visualization and equipped with multi-chips by the development and injection of a highly stiff bone-like 3D matrix, made of a blend of collagen-enriched natural hydrogels loaded with hydroxyapatite nanocrystals. The microchannel, filled with the ostemimetic matrix and Oc, is subjected to passive perfusion and shear stress. We used scanning electron microscopy for preliminary material characterization. Confocal microscopy and fluorescent microbeads were used after material injection into the microchannels to detect volume changes and the distribution of cell-sized objects within the hydrogel. The formation of a 3D dendritic network of Oc was monitored by measuring cell viability, evaluating phenotyping markers (connexin43, integrin alpha V/CD51, sclerostin), quantification of dendrites, and responsiveness to an anabolic drug. The platform is expected to accelerate the development of new drug aimed at modulating the survival and function of osteocytes.
Collapse
Affiliation(s)
| | - Gemma Di Pompo
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Boanini
- Department of Chemistry 'Giacomo Ciamician', University of Bologna, Bologna, Italy
| | - Gabriela Graziani
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Enrico Sassoni
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Roles of miR-196a and miR-196b in Zebrafish Motor Function. Biomolecules 2023; 13:biom13030554. [PMID: 36979489 PMCID: PMC10046552 DOI: 10.3390/biom13030554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Background: The exertion of motor function depends on various tissues, such as bones and muscles. miR-196 has been widely studied in cancer and other fields, but its effect on bone and skeletal muscle is rarely reported. In order to explore the role of miR-196 family in bone and skeletal muscle, we used the previously successfully constructed miR-196a-1 and miR-196b gene knockout zebrafish animal models for research. Methods: The behavioral trajectories of zebrafish from 4 days post-fertilization (dpf) to 7 dpf were detected to analyze the effect of miR-196a-1 and miR-196b on motor ability. Hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) were used to detect the dorsal muscle tissue of zebrafish. The bone tissue of zebrafish was detected by microcomputed tomography (micro-CT). Real-time PCR was used to detect the expression levels of related genes, including vcp, dpm1, acta1b, mylpfb, col1a1a, bmp8a, gdf6a, and fgfr3. Results: The behavioral test showed that the total behavioral trajectory, movement time, and movement speed of zebrafish larvae were decreased in the miR-196a-1 and miR-196b gene knockout lines. Muscle tissue analysis showed that the structure of muscle fibers in the zebrafish lacking miR-196a-1 and miR-196b was abnormal and was characterized by vacuolar degeneration of muscle fibers, intranuclear migration, melanin deposition, and inflammatory cell infiltration. Bone CT examination revealed decreased bone mineral density and trabecular bone number. The real-time PCR results showed that the expression levels of vcp, dpm1, gdf6a, fgfr3, and col1a1a were decreased in the miR-196b gene knockout group. The expression levels of dpm1, acta1b, mylpfb, gdf6a, and col1a1a were decreased, and the expression level of fgfr3 was increased in the miR-196b gene knockout group compared with the wild-type group. Conclusions: miR-196a-1 and miR-196b play an important role in muscle fiber structure, bone mineral density, and bone trabecular quantity by affecting the expression of vcp, dpm1, acta1b, mylpfb, gdf6a, fgfr3, and col1a1a and then affect the function of the motor system
Collapse
|
15
|
Maciel GBM, Maciel RM, Danesi CC. Bone cells and their role in physiological remodeling. Mol Biol Rep 2023; 50:2857-2863. [PMID: 36609750 DOI: 10.1007/s11033-022-08190-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE This work compiles the characteristics of bone cells involved in the physiological bone remodeling. METHODS A narrative review of the literature was performed. RESULTS Remodeling is a different process from modeling. Remodeling allows old or damaged bone tissue to be renewed, ensuring the maintenance of bone fracture resistance, as well as maintaining calcium and phosphorus homeostasis. We present the role of osteoclasts, a multinucleated cell with hematopoietic origin responsible for resorbing bone. The formation of osteoclasts depends on the cytokines macrophage colony stimulating factor (M-CSF) and receptor activator of NF-kB ligand (RANKL) and can be blocked by osteoprotegerin. Furthermore, this review highlights the features of osteoblasts, polarized cubic cells of mesenchymal origin that deposit bone and also covers osteocytes and bone lining cells. This review presents the five fundamental phases of bone remodeling and addresses aspects of its regulation through hormones and growth factors. CONCLUSIONS Knowledge of the current concepts of physiological bone remodeling is necessary for the study of the different pathologies that affect the bone tissue and thus helps in the search for new therapies.
Collapse
Affiliation(s)
- Gabriel Bassan Marinho Maciel
- Postgraduate Program in Dental Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil. .,Department of Pathology, Federal University of Santa Maria, Av. Roraima, 1000, Santa Maria, 97015-900, RS, Brazil.
| | | | | |
Collapse
|
16
|
Blouin S, Misof BM, Mähr M, Fratzl-Zelman N, Roschger P, Lueger S, Messmer P, Keplinger P, Rauch F, Glorieux FH, Berzlanovich A, Gruber GM, Brugger PC, Shane E, Recker RR, Zwerina J, Hartmann MA. Osteocyte lacunae in transiliac bone biopsy samples across life span. Acta Biomater 2023; 157:275-287. [PMID: 36549635 DOI: 10.1016/j.actbio.2022.11.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022]
Abstract
Osteocytes act as bone mechanosensors, regulators of osteoblast/osteoclast activity and mineral homeostasis, however, knowledge about their functional/morphological changes throughout life is limited. We used quantitative backscattered electron imaging (qBEI) to investigate osteocyte lacunae sections (OLS) as a 2D-surrogate characterizing the osteocytes. OLS characteristics, the density of mineralized osteocyte lacunae (i.e., micropetrotic osteocytes, md.OLS-Density in nb/mm2) and the average degree of mineralization (CaMean in weight% calcium) of cortex and spongiosa were analyzed in transiliac biopsy samples from healthy individuals under 30 (n=59) and over 30 years (n=50) (i.e., before and after the age of peak bone mass, respectively). We found several differences in OLS-characteristics: 1). Inter-individually between the age groups: OLS-Density and OLS-Porosity were reduced by about 20% in older individuals in spongiosa and in cortex versus younger probands (both, p < 0.001). 2). Intra-individually between bone compartments: OLS-Density was higher in the cortex, +18.4%, p < 0.001 for younger and +7.6%, p < 0.05 for older individuals. Strikingly, the most frequent OLS nearest-neighbor distance was about 30 µm in both age groups and at both bone sites revealing a preferential organization of osteocytes in clusters. OLS-Density was negatively correlated with CaMean in both spongiosa and cortex (both, p < 0.001). Few mineralized OLS were found in young individuals along with an increase of md.OLS-Density with age. In summary, this transiliac bone sample analysis of 200000 OLS from 109 healthy individuals throughout lifespan reveals several age-related differences in OLS characteristics. Moreover, our study provides reference data from healthy individuals for different ages to be used for diagnosis of bone abnormalities in diseases. STATEMENT OF SIGNIFICANCE: Osteocytes are bone cells embedded in lacunae within the mineralized bone matrix and have a key role in the bone metabolism and the mineral homeostasis. Not easily accessible, we used quantitative backscattered electron imaging to determine precisely number and shape descriptors of the osteocyte lacunae in 2D. We analyzed transiliac biopsy samples from 109 individuals with age distributed from 2 to 95 years. Compact cortical bone showed constantly higher lacunar density than cancellous bone but the lacunar density in both bone tissue decreased with age before the peak bone mass age at 30 years and stabilized or even increased after this age. This extensive study provides osteocyte lacunae reference data from healthy individuals usable for bone pathology diagnosis.
Collapse
Affiliation(s)
- Stéphane Blouin
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria.
| | - Barbara M Misof
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Matthias Mähr
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Paul Roschger
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Sonja Lueger
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Phaedra Messmer
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Petra Keplinger
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Frank Rauch
- Shriners Hospital for Children and McGill University, Montreal, ON QC, H4A 0A9, Canada
| | - Francis H Glorieux
- Shriners Hospital for Children and McGill University, Montreal, ON QC, H4A 0A9, Canada
| | - Andrea Berzlanovich
- Unit of Forensic Gerontology, Center of Forensic Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerlinde M Gruber
- Department of Anatomy and Biomechanics, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Peter C Brugger
- Center for Anatomy and Cell Biology, Department of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Elizabeth Shane
- Department of Medicine, Division of Endocrinology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Robert R Recker
- Osteoporosis Research Center, Creighton University, Omaha, Nebraska, USA
| | - Jochen Zwerina
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria & Vienna Bone and Growth Center, Vienna, Austria
| |
Collapse
|
17
|
Osteocytic Sclerostin Expression as an Indicator of Altered Bone Turnover. Nutrients 2023; 15:nu15030598. [PMID: 36771305 PMCID: PMC9921466 DOI: 10.3390/nu15030598] [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: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
Renal osteodystrophy (ROD) is a complex and serious complication of chronic kidney disease (CKD), a major global health problem caused by loss of renal function. Currently, the gold standard to accurately diagnose ROD is based on quantitative histomorphometric analysis of trabecular bone. Although this analysis encompasses the evaluation of osteoblast and osteoclast number/activity, tfigurehe interest in osteocytes remains almost nihil. Nevertheless, this cell type is evidenced to perform a key role in bone turnover, particularly through its production of various bone proteins, such as sclerostin. In this study, we aim to investigate, in the context of ROD, to which extent an association exists between bone turnover and the abundance of osteocytes and osteocytic sclerostin expression in both the trabecular and cortical bone compartments. Additionally, the effect of parathyroid hormone (PTH) on bone sclerostin expression was examined in parathyroidectomized rats. Our results indicate that PTH exerts a direct inhibitory function on sclerostin, which in turn negatively affects bone turnover and mineralization. Moreover, this study emphasizes the functional differences between cortical and trabecular bone, as the number of (sclerostin-positive) osteocytes is dependent on the respective bone compartment. Finally, we evaluated the potential of sclerostin as a marker for CKD and found that the diagnostic performance of circulating sclerostin is limited and that changes in skeletal sclerostin expression occur more rapidly and more pronounced. The inclusion of osteocytic sclerostin expression and cortical bone analysis could be relevant when performing bone histomorphometric analysis for diagnostic purposes and to unravel pathological mechanisms of bone disease.
Collapse
|
18
|
Liu X, Hu J, Ning Y, Xu H, Cai H, Yang A, Shi Z, Li Z. Aptamer Technology and Its Applications in Bone Diseases. Cell Transplant 2023; 32:9636897221144949. [PMID: 36591965 PMCID: PMC9811309 DOI: 10.1177/09636897221144949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aptamers are single-stranded nucleic acids (DNA, short RNA, or other artificial molecules) produced by the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) technology, which can be tightly and specifically combined with desired targets. As a comparable alternative to antibodies, aptamers have many advantages over traditional antibodies such as a strong chemical stability and rapid bulk production. In addition, aptamers can bind targets in various ways, and are not limited like the antigen-antibody combination. Studies have shown that aptamers have tremendous potential to diagnose and treat clinical diseases. However, only a few aptamer-based drugs have been used because of limitations of the aptamers and SELEX technology. To promote the development and applications of aptamers, we present a review of the methods optimizing the SELEX technology and modifying aptamers to boost the selection success rate and improve aptamer characteristics. In addition, we review the application of aptamers to treat bone diseases.
Collapse
Affiliation(s)
- Xiangzhong Liu
- Department of Orthopaedics, Wuhan Third
Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Jing Hu
- Wuhan Children’s Hospital, Tongji
Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yu Ning
- Department of Orthopaedics, Xiangyang
Hospital of Traditional Chinese Medicine Affiliated to Hubei University of Chinese
Medicine, Xiangyang, China
| | - Haijia Xu
- Department of Orthopaedics, Wuhan Third
Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Hantao Cai
- Department of Orthopaedics, Wenling
First People’s Hospital, Taizhou, China
| | - Aofei Yang
- Department of Orthopaedics, Hubei
Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Zhengshuai Shi
- Department of Orthopaedics, Wuhan
Sports University, Wuhan, China
| | - Zhanghua Li
- Department of Orthopaedics, Wuhan Third
Hospital, Tongren Hospital of Wuhan University, Wuhan, China,Zhanghua Li, Department of Orthopaedics,
Wuhan Third Hospital, Tongren Hospital of Wuhan University, No. 216, Guanshan
Avenue, Hongshan District, Wuhan 430074, Hubei Province, China.
| |
Collapse
|
19
|
Impact of X-Linked Hypophosphatemia on Muscle Symptoms. Genes (Basel) 2022; 13:genes13122415. [PMID: 36553684 PMCID: PMC9778127 DOI: 10.3390/genes13122415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
X-linked hypophosphatemia (XLH) is the most common hereditary form of rickets and deficiency of renal tubular phosphate transport in humans. XLH is caused by the inactivation of mutations within the phosphate-regulating endopeptidase homolog X-linked (PHEX) gene and follows an X-dominant transmission. It has an estimated frequency of 1 case per 20,000, and over 300 distinct pathogenic variations have been reported that result in an excess of fibroblast growth factor 23 (FGF23) in the serum. Increased levels of FGF23 lead to renal phosphate loss, decreased serum 1,25-dihydroxyvitamin D, and increased metabolism of 1,25-dihydoxyvitamin D, resulting in hypophosphatemia. Major clinical manifestations include rickets, bone deformities, and growth retardation that develop during childhood, and osteomalacia-related fractures or pseudo-fractures, degenerative osteoarthritis, enthesopathy, dental anomalies, and hearing loss during adulthood, which can affect quality of life. In addition, fatigue is also a common symptom in patients with XLH, who experience decreased motion, muscle weakness, and pain, contributing to altered quality of life. The clinical and biomedical characteristics of XLH are extensively defined in bone tissue since skeletal deformations and mineralization defects are the most evident effects of high FGF23 and low serum phosphate levels. However, despite the muscular symptoms that XLH causes, very few reports are available on the effects of FGF23 and phosphate in muscle tissue. Given the close relationship between bones and skeletal muscles, studying the effects of FGF23 and phosphate on muscle could provide additional opportunities to understand the interactions between these two important compartments of the body. By describing the current literature on XLH and skeletal muscle dysfunctions, the purpose of this review is to highlight future areas of research that could contribute to a better understanding of XLH muscular disability and its management.
Collapse
|
20
|
Differential Expression of Dickkopf 1 and Periostin in Mouse Strains with High and Low Bone Mass. BIOLOGY 2022; 11:biology11121840. [PMID: 36552348 PMCID: PMC9775221 DOI: 10.3390/biology11121840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/01/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
By expressing different genes and proteins that regulate osteoclast as well as osteoblast formation, osteocytes orchestrate bone metabolism. The aim of this project was the evaluation of the differences in the osteocytes’ secretory activity in the low bone mass mouse strain C57BL/6J and the high bone mass strain C3H/J. The femura of eight- and sixteen-week-old male C57BL/6J and C3H/J mice—six animals per group—were analyzed. Using immunohistochemistry, osteocytes expressing dickkopf 1, sclerostin, periostin, fibroblast growth factor 23 (FGF23), and osteoprotegerin were detected. By means of the OsteoMeasure-System, 92.173 osteocytes were counted. At the age of eight weeks, approximately twice as many cortical and trabecular osteocytes from the C57BL/6J mice compared to the C3H/J mice expressed dickkopf 1 (p < 0.005). The number of cortical osteocytes expressing sclerostin was also higher in the C57BL/6J mice (p < 0.05). In contrast, the cortical and trabecular osteocytes expressing periostin were twice as high in the C3H/J mice (p < 0.005). The dickkopf 1 expressing osteocytes of the C57BL/6J mice decreased with age and showed a strain-specific difference only in cortical bone by 16 weeks of age (p < 0.05). In the C3H/J mice, the amount of osteocytes expressing periostin tended to increase with age. Thus, strain-related differences were maintained in 16-week-old rodents (p < 0.005). No strain-specific differences in the expression of FGF23 or osteoprotegerin in the cortical compartment could be detected. This experimental study showed that the osteocytes’ protein expression reflects differences in bone characteristics and strain-related differences during skeletal maturation. Besides the osteocytes’ expression of sclerostin, their expression of dickkopf 1 and periostin seems to be important for bone properties as well.
Collapse
|
21
|
Zhang Y, Dong F, Wang Z, Xu B, Zhang T, Wang Q, Lin Q. Fluoride Exposure Provokes Mitochondria-Mediated Apoptosis and Increases Mitophagy in Osteocytes via Increasing ROS Production. Biol Trace Elem Res 2022:10.1007/s12011-022-03450-w. [PMID: 36255553 DOI: 10.1007/s12011-022-03450-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/13/2022] [Indexed: 11/28/2022]
Abstract
Fluoride is a persistent environmental pollutant, and its excessive intake causes skeletal and dental fluorosis. However, few studies focused on the effects of fluoride on osteocytes, making up over 95% of all bone cells. This study aimed to investigate the effect of fluoride on osteocytes in vitro, as well as explore the underlying mechanisms. CCK-8, LDH assay, fluorescent probes, flow cytometry, and western blotting were performed to examine cell viability, apoptosis, mitochondria changes, reactive oxygen species (ROS) and mitochondrial ROS (mtROS), and protein expressions. Results showed that sodium fluoride (NaF) exposure (4, 8 mmol/L) for 24 h inhibited the cell viability of osteocytes MLO-Y4 and promoted G0/G1 phase arrest and increased cell apoptosis. NaF treatment remarkably caused mitochondria damage, loss of MMP, ATP decrease, Cyto c release, and Bax/Bcl-2 ratio increase and elevated the activity of caspase-9 and caspase-3. Furthermore, NaF significantly upregulated the expressions of LC-3II, PINK1, and Parkin and increased autophagy flux and the accumulation of acidic vacuoles, while the p62 level was downregulated. In addition, NaF exposure triggered the production of intracellular ROS and mtROS and increased malondialdehyde (MDA); but superoxide dismutase (SOD) activity and glutathione (GSH) content were decreased. The scavenger N-acetyl-L-cysteine (NAC) significantly reversed NaF-induced apoptosis and mitophagy, suggesting that ROS is responsible for the mitochondrial-mediated apoptosis and mitophagy induced by NaF exposure. These findings provide in vitro evidence that apoptosis and mitophagy are cellular mechanisms for the toxic effect of fluoride on osteocytes, thereby suggesting the potential role of osteocytes in skeletal and dental fluorosis.
Collapse
Affiliation(s)
- Yun Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China.
| | - Fanhe Dong
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Zihan Wang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Bingbing Xu
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Tao Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Qiqi Wang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Qiao Lin
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| |
Collapse
|
22
|
Imamudeen N, Basheer A, Iqbal AM, Manjila N, Haroon NN, Manjila S. Management of Osteoporosis and Spinal Fractures: Contemporary Guidelines and Evolving Paradigms. Clin Med Res 2022; 20:95-106. [PMID: 35478096 PMCID: PMC9242734 DOI: 10.3121/cmr.2021.1612] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/17/2021] [Indexed: 01/24/2023]
Abstract
Physicians involved in treating spine fractures secondary to osteopenia and osteoporosis should know the pathogenesis and current guidelines on managing the underlying diminished bone mineral density, as worldwide fracture prevention campaigns are trailing behind in meeting their goals. This is a narrative review exploring the various imaging and laboratory tests used to diagnose osteoporotic fractures and a comprehensive compilation of contemporary medical and surgical management. We have incorporated salient recommendations from the Endocrine Society, the American Association of Clinical Endocrinology (AACE), and the American Society for Bone and Mineral Research (ASBMR). The use of modern scoring systems such as Fracture Risk Assessment Tool (FRAX®) for evaluating fracture risk in osteoporosis with a 10-year probability of hip fracture and major fractures in the spine, forearm, hip, or shoulder is highlighted. This osteoporosis risk assessment tool can be easily incorporated into the preoperative bone health optimization strategies, especially before elective spine surgery in osteoporotic patients. The role of primary surgical intervention for vertebral compression fracture and secondary fracture prevention with pharmacological therapy is described, with randomized clinical trial-based wisdom on its timing and dosage, drug holiday, adverse effects, and relevant evidence-based literature. We also aim to present an evidence-based clinical management algorithm for treating osteoporotic vertebral body compression fractures, tumor-induced osteoporosis, or hardware stabilization in elderly trauma patients in the setting of their impaired bone health. The recent guidelines and recommendations on surgical intervention by various medical societies are covered, along with outcome studies that reveal the efficacy of cement augmentation of vertebral compression fractures via vertebroplasty and balloon kyphoplasty versus conservative medical management in the elderly population.
Collapse
Affiliation(s)
- Nasvin Imamudeen
- Department of Medicine, Marshfield Medical Center, Marshfield, Wisconsin, USA
| | - Amjad Basheer
- Department of Medicine, University of Connecticut, CT, USA
| | - Anoop Mohamed Iqbal
- Division of Pediatric Endocrinology, Marshfield Medical Center, Marshfield, Wisconsin, USA
| | - Nihal Manjila
- Department of History and Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nisha Nigil Haroon
- Department of Neurosurgery, Ayer Neuroscience Institute, The Hospital of Central Connecticut, New Britain, Connecticut, USA
| | - Sunil Manjila
- Division of Pediatric Endocrinology, Marshfield Medical Center, Marshfield, Wisconsin, USA
| |
Collapse
|
23
|
Choi JUA, Kijas AW, Lauko J, Rowan AE. The Mechanosensory Role of Osteocytes and Implications for Bone Health and Disease States. Front Cell Dev Biol 2022; 9:770143. [PMID: 35265628 PMCID: PMC8900535 DOI: 10.3389/fcell.2021.770143] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Bone homeostasis is a dynamic equilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts. This process is primarily controlled by the most abundant and mechanosensitive bone cells, osteocytes, that reside individually, within chambers of porous hydroxyapatite bone matrix. Recent studies have unveiled additional functional roles for osteocytes in directly contributing to local matrix regulation as well as systemic roles through endocrine functions by communicating with distant organs such as the kidney. Osteocyte function is governed largely by both biochemical signaling and the mechanical stimuli exerted on bone. Mechanical stimulation is required to maintain bone health whilst aging and reduced level of loading are known to result in bone loss. To date, both in vivo and in vitro approaches have been established to answer important questions such as the effect of mechanical stimuli, the mechanosensors involved, and the mechanosensitive signaling pathways in osteocytes. However, our understanding of osteocyte mechanotransduction has been limited due to the technical challenges of working with these cells since they are individually embedded within the hard hydroxyapatite bone matrix. This review highlights the current knowledge of the osteocyte functional role in maintaining bone health and the key regulatory pathways of these mechanosensitive cells. Finally, we elaborate on the current therapeutic opportunities offered by existing treatments and the potential for targeting osteocyte-directed signaling.
Collapse
Affiliation(s)
- Jung Un Ally Choi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda W Kijas
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Jan Lauko
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Alan E Rowan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
25
|
Domazetovic V, Falsetti I, Ciuffi S, Iantomasi T, Marcucci G, Vincenzini MT, Brandi ML. Effect of Oxidative Stress-Induced Apoptosis on Active FGF23 Levels in MLO-Y4 Cells: The Protective Role of 17-β-Estradiol. Int J Mol Sci 2022; 23:ijms23042103. [PMID: 35216216 PMCID: PMC8879671 DOI: 10.3390/ijms23042103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/04/2022] Open
Abstract
The discovery that osteocytes secrete phosphaturic fibroblast growth factor 23 (FGF23) has defined bone as an endocrine organ. However, the autocrine and paracrine functions of FGF23 are still unknown. The present study focuses on the cellular and molecular mechanisms involved in the complex control of FGF23 production and local bone remodeling functions. FGF23 was assayed using ELISA kit in the presence or absence of 17β–estradiol in starved MLO-Y4 osteocytes. In these cells, a relationship between oxidative stress-induced apoptosis and up-regulation of active FGF23 levels due to MAP Kinases activation with involvement of the transcriptional factor (NF-kB) has been demonstrated. The active FGF23 increase can be due to up-regulation of its expression and post-transcriptional modifications. 17β–estradiol prevents the increase of FGF23 by inhibiting JNK and NF-kB activation, osteocyte apoptosis and by the down-regulation of osteoclastogenic factors, such as sclerostin. No alteration in the levels of dentin matrix protein 1, a FGF23 negative regulator, has been determined. The results of this study identify biological targets on which drugs and estrogen may act to control active FGF23 levels in oxidative stress-related bone and non-bone inflammatory diseases.
Collapse
Affiliation(s)
- Vladana Domazetovic
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Simone Ciuffi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Gemma Marcucci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Maria Teresa Vincenzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (V.D.); (I.F.); (S.C.); (T.I.); (G.M.); (M.T.V.)
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
26
|
Coşkun Benlidayı İ. The Osteocyte as a Director of Bone Metabolism. Arch Rheumatol 2021; 36:617-619. [PMID: 35382360 PMCID: PMC8957769 DOI: 10.46497/archrheumatol.2021.8632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/28/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- İlke Coşkun Benlidayı
- Department of Physical Medicine and Rehabilitation, Çukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
27
|
Hariri H, St-Arnaud R. Expression and Role of Ubiquitin-Specific Peptidases in Osteoblasts. Int J Mol Sci 2021; 22:ijms22147746. [PMID: 34299363 PMCID: PMC8304380 DOI: 10.3390/ijms22147746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 11/16/2022] Open
Abstract
The ubiquitin-proteasome system regulates biological processes in normal and diseased states. Recent investigations have focused on ubiquitin-dependent modifications and their impacts on cellular function, commitment, and differentiation. Ubiquitination is reversed by deubiquitinases, including ubiquitin-specific peptidases (USPs), whose roles have been widely investigated. In this review, we explore recent findings highlighting the regulatory functions of USPs in osteoblasts and providing insight into the molecular mechanisms governing their actions during bone formation. We also give a brief overview of our work on USP53, a target of PTH in osteoblasts and a regulator of mesenchymal cell lineage fate decisions. Emerging evidence addresses questions pertaining to the complex layers of regulation exerted by USPs on osteoblast signaling. We provide a short overview of our and others' understanding of how USPs modulate osteoblastogenesis. However, further studies using knockout mouse models are needed to fully understand the mechanisms underpinning USPs actions.
Collapse
Affiliation(s)
- Hadla Hariri
- Research Centre, Shriners Hospital for Children, Montreal, QC H4A 0A9, Canada;
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - René St-Arnaud
- Research Centre, Shriners Hospital for Children, Montreal, QC H4A 0A9, Canada;
- Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 1A4, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 1A1, Canada
- Correspondence: ; Tel.: +514-282-7155; Fax: +514-842-5581
| |
Collapse
|
28
|
Wang YY, Xia K, Wang ZX, Xie H, Xu R. Osteocyte exosomes accelerate benign prostatic hyperplasia development. Mol Cell Endocrinol 2021; 531:111301. [PMID: 33933560 DOI: 10.1016/j.mce.2021.111301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/05/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
Benign prostatic hyperplasia (BPH) is one of the most common diseases in elderly men. BPH patients exhibit an increased risk of vertebral and hip fractures, which are most attributable to pre-existing osteoporosis. However, the relationship between BPH and osteoporosis is still unknown. Here we found that osteocytes, the most abundant bone cells, promoted BPH development by secreting exosomes. In vitro, osteocyte exosomes (OCY-Exo) directly promoted cell proliferation of a prostate epithelial cell line BPH-1 and a macrophage cell line RAW264.7, OCY-Exo also stimulated macrophage-induced proliferation of BPH-1 cells. In vivo, intramedullary injection of OCY-Exo accumulated in prostate. Intravenous administration of OCY-Exo exacerbated testosterone-induced BPH in C57BL/6J mice. Our study uncovers the role of OCY-Exo as a stimulator of BPH, suggesting a novel mechanism in bone-prostate communication.
Collapse
Affiliation(s)
- Yi-Yi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
29
|
Canter BS, Leung CN, Fritton JC, Bäck T, Rajon D, Azzam EI, Howell RW. Radium-223-induced Bystander Effects Cause DNA Damage and Apoptosis in Disseminated Tumor Cells in Bone Marrow. Mol Cancer Res 2021; 19:1739-1750. [PMID: 34039648 DOI: 10.1158/1541-7786.mcr-21-0005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/02/2021] [Accepted: 05/19/2021] [Indexed: 11/16/2022]
Abstract
Radiation-induced bystander effects have been implicated in contributing to the growth delay of disseminated tumor cells (DTC) caused by 223RaCl2, an alpha particle-emitting radiopharmaceutical. To understand how 223RaCl2 affects the growth, we have quantified biological changes caused by direct effects of radiation and bystander effects caused by the emitted radiations on DTC and osteocytes. Characterizing these effects contribute to understanding the efficacy of alpha particle-emitting radiopharmaceuticals and guide expansion of their use clinically. MDA-MB-231 or MCF-7 human breast cancer cells were inoculated intratibially into nude mice that were previously injected intravenously with 50 or 600 kBq/kg 223RaCl2. At 1-day and 3-days postinoculation, tibiae were harvested and examined for DNA damage (γ-H2AX foci) and apoptosis in osteocytes and cancer cells located within and beyond the range (70 μm) of alpha particles emitted from the bone surface. Irradiated and bystander MDA-MB-231 and MCF-7 cells harbored DNA damage. Bystander MDA-MB-231 cells expressed DNA damage at both treatment levels while bystander MCF-7 cells required the higher administered activity. Osteocytes also had DNA damage regardless of inoculated cancer cell line. The extent of DNA damage was quantified by increases in low (1-2 foci), medium (3-5 foci), and high (5+ foci) damage. MDA-MB-231 but not MCF-7 bystander cells showed increases in apoptosis in 223RaCl2-treated animals, as did irradiated osteocytes. In summary, radiation-induced bystander effects contribute to DTC cytotoxicity caused by 223RaCl2. IMPLICATIONS: This observation supports clinical investigation of the efficacy of 223RaCl2 to prevent breast cancer DTC from progressing to oligometastases.
Collapse
Affiliation(s)
- Brian S Canter
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Calvin N Leung
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - J Christopher Fritton
- Departments of Mechanical and Biomedical Engineering, City College of New York, New York, New York
| | - Tom Bäck
- Department of Radiation Physics, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Didier Rajon
- Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Edouard I Azzam
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey.,Radiobiology and Health Branch, Canadian Nuclear Laboratories, Ontario, Canada
| | - Roger W Howell
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey.
| |
Collapse
|
30
|
Gerosa L, Lombardi G. Bone-to-Brain: A Round Trip in the Adaptation to Mechanical Stimuli. Front Physiol 2021; 12:623893. [PMID: 33995117 PMCID: PMC8120436 DOI: 10.3389/fphys.2021.623893] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Besides the classical ones (support/protection, hematopoiesis, storage for calcium, and phosphate) multiple roles emerged for bone tissue, definitively making it an organ. Particularly, the endocrine function, and in more general terms, the capability to sense and integrate different stimuli and to send signals to other tissues, has highlighted the importance of bone in homeostasis. Bone is highly innervated and hosts all nervous system branches; bone cells are sensitive to most of neurotransmitters, neuropeptides, and neurohormones that directly affect their metabolic activity and sensitivity to mechanical stimuli. Indeed, bone is the principal mechanosensitive organ. Thanks to the mechanosensing resident cells, and particularly osteocytes, mechanical stimulation induces metabolic responses in bone forming (osteoblasts) and bone resorbing (osteoclasts) cells that allow the adaptation of the affected bony segment to the changing environment. Once stimulated, bone cells express and secrete, or liberate from the entrapping matrix, several mediators (osteokines) that induce responses on distant targets. Brain is a target of some of these mediator [e.g., osteocalcin, lipocalin2, sclerostin, Dickkopf-related protein 1 (Dkk1), and fibroblast growth factor 23], as most of them can cross the blood-brain barrier. For others, a role in brain has been hypothesized, but not yet demonstrated. As exercise effectively modifies the release and the circulating levels of these osteokines, it has been hypothesized that some of the beneficial effects of exercise on brain functions may be associated to such a bone-to-brain communication. This hypothesis hides an interesting clinical clue: may well-addressed physical activities support the treatment of neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases?
Collapse
Affiliation(s)
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
31
|
Ofer L, Zaslansky P, Shahar R. A comparison of the structure, composition and mechanical properties of anosteocytic vertebrae of medaka (O. latipes) and osteocytic vertebrae of zebrafish (D. rerio). JOURNAL OF FISH BIOLOGY 2021; 98:995-1006. [PMID: 32239680 DOI: 10.1111/jfb.14334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 03/06/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Medaka (O. latipes) and zebrafish (D. rerio) are two teleost fish increasingly used as models to study human skeletal diseases. Although they are similar in size, swimming pattern and many other characteristics, these two species are very distant from an evolutionary point of view (by at least 100 million years). A prominent difference between the skeletons of medaka and zebrafish is the total absence of osteocytes in medaka (anosteocytic), while zebrafish bone contains numerous osteocytes (osteocytic). This fundamental difference suggests the possibility that the bony elements of their skeleton may be different in a variety of other aspects, structural, mechanical or both, particularly in heavily loaded bones like the vertebrae. Here we report on the results of a comparative study that aimed to determine the similarities and differences in medaka and zebrafish vertebrae in terms of their macro- to nanostructure, composition and mechanical properties. Our results reveal many similarities between medaka and zebrafish vertebrae, making the lack or presence of osteocytes the only major difference between the bones of these two species.
Collapse
Affiliation(s)
- Lior Ofer
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Paul Zaslansky
- Department for Restorative and Preventive Dentistry, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Ron Shahar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
32
|
Wang Y, Xu L, Wang J, Bai J, Zhai J, Zhu G. Radiation induces primary osteocyte senescence phenotype and affects osteoclastogenesis in vitro. Int J Mol Med 2021; 47:76. [PMID: 33693957 PMCID: PMC7949628 DOI: 10.3892/ijmm.2021.4909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Irradiation-induced bone remodeling imbalances arise as a consequence of the dysregulation of bone formation and resorption. Due to the abundance of osteocytes, their long life and their dual-regulatory effects on both osteoblast and osteoclast function, they serve as critical coordinators of bone remolding. In the present study, femur and tibia-derived primary osteocytes were cultured and irradiated to observe the functional changes and the cellular senescence phenotype in vitro. Irradiation directly reduced cell viability, affected the crucial dendritic morphology and altered the expression of functional proteins, including upregulation of receptor activator of nuclear factor-κB ligand and sclerostin, and downregulation of osteoprotegerin. Irradiated osteocytes were shown to exhibit notable DNA damage, which resulted in the initiation of a typical cellular senescence phenotype. Furthermore, it was found that irradiation-induced prematurely senescent osteocytes stimulate molecular secretion, referred to as senescence-associated secretory phenotype (SASP), which may be involved in modulation of the bone microenvironment, including the promotion of osteoclastogenesis. Taken together, the results showed that irradiation triggered osteocyte senescence and the acquisition of an associated secretory phenotype. This further resulted in an imbalance of bone remodeling through senescent influence on proliferation, morphology and marker protein production, but also indirectly via a paracrine pathway through SASP secretion. The results of the present study may highlight the potential of SASP-targeted interventions for the management of radiation-induced bone loss.
Collapse
Affiliation(s)
- Yuyang Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Linshan Xu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianping Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jiangtao Bai
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianglong Zhai
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Guoying Zhu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
33
|
Nguyen HD, Sun X, Yokota H, Lin CC. Probing Osteocyte Functions in Gelatin Hydrogels with Tunable Viscoelasticity. Biomacromolecules 2021; 22:1115-1126. [PMID: 33543929 PMCID: PMC10548335 DOI: 10.1021/acs.biomac.0c01476] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bone is an attractive site for metastatic cancer cells and has been considered as "soil" for promoting tumor growth. However, accumulating evidence suggests that some bone cells (e.g., osteocytes) can actually suppress cancer cell migration and invasion via direct cell-cell contact and/or through cytokine secretion. Toward designing a biomimetic niche for supporting 3D osteocyte culture, we present here a gelatin-based hydrogel system with independently tunable matrix stiffness and viscoelasticity. In particular, we synthesized a bifunctional macromer, gelatin-norbornene-boronic acid (i.e., GelNB-BA), for covalent cross-linking with multifunctional thiol linkers [e.g., four-arm poly(ethylene glycol)-thiol or PEG4SH] to form thiol-NB hydrogels. The immobilized BA moieties in the hydrogel readily formed reversible boronate ester bonds with 1,3-diols on physically entrapped poly(vinyl alcohol) (PVA). Adjusting the compositions of GelNB-BA, PEG4SH, and PVA afforded hydrogels with independently tunable elasticity and viscoelasticity. With this new dynamic hydrogel platform, we investigated matrix mechanics-induced growth and cytokine secretion of encapsulated MLO-A5 pre-osteocytes. We discovered that more compliant or viscoelastic gels promoted A5 cell growth. On the other hand, cells encapsulated in stiffer gels secreted higher amounts of pro-inflammatory cytokines and chemokines. Finally, conditioned media (CM) collected from the encapsulated MLO-A5 cells (i.e., A5-CM) strongly inhibited breast cancer cell proliferation, invasion, and expression of tumor-activating genes. This new biomimetic hydrogel platform not only serves as a versatile matrix for investigating mechano-sensing in osteocytes but also provides a means to produce powerful anti-tumor CM.
Collapse
Affiliation(s)
- Han D. Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Xun Sun
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Hiroki Yokota
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
34
|
Goel D, Vohora D. Liver X receptors and skeleton: Current state-of-knowledge. Bone 2021; 144:115807. [PMID: 33333244 DOI: 10.1016/j.bone.2020.115807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/25/2022]
Abstract
The liver X receptors (LXR) is a nuclear receptor that acts as a prominent regulator of lipid homeostasis and inflammatory response. Its therapeutic effectiveness against various diseases like Alzheimer's disease and atherosclerosis has been investigated in detail. Emerging pieces of evidence now reveal that LXR is also a crucial modulator of bone remodeling. However, the molecular mechanisms underlying the pharmacological actions of LXR on the skeleton and its role in osteoporosis are poorly understood. Therefore, in the current review, we highlight LXR and its actions through different molecular pathways modulating skeletal homeostasis. The studies described in this review propound that LXR in association with estrogen, PTH, PPARγ, RXR hedgehog, and canonical Wnt signaling regulates osteoclastogenesis and bone resorption. It regulates RANKL-induced expression of c-Fos, NFATc1, and NF-κB involved in osteoclast differentiation. Additionally, several studies suggest suppression of RANKL-induced osteoclast differentiation by synthetic LXR ligands. Given the significance of modulation of LXR in various physiological and pathological settings, our findings indicate that therapeutic targeting of LXR might potentially prevent or treat osteoporosis and improve bone quality.
Collapse
Affiliation(s)
- Divya Goel
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
35
|
Abstract
MicroRNAs (miRNAs) are a class of short RNA molecules that mediate the regulation of gene activity through interactions with target mRNAs and subsequent silencing of gene expression. It has become increasingly clear the miRNAs regulate many diverse aspects of bone biology, including bone formation and bone resorption processes. The role of miRNAs specifically in osteoclasts has been of recent investigation, due to clinical interest in discovering new paradigms to control excessive bone resorption, as is observed in multiple conditions including aging, estrogen deprivation, cancer metastases or glucocorticoid use. Therefore understanding the role that miRNAs play during osteoclastic differentiation is of critical importance. In this review, we highlight and discuss general aspects of miRNA function in osteoclasts, including exciting data demonstrating that miRNAs encapsulated in extracellular vesicles (EVs) either originating from osteoclasts, or signaling to osteoclast from divergent sites, have important roles in bone homeostasis.
Collapse
Affiliation(s)
- Megan M Weivoda
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Sun-Kyeong Lee
- Department of Medicine, UCONN Center on Aging, University Connecticut Health Center, Farmington, CT 06030, USA
| | - David G Monroe
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
Abstract
Development of cartilage and bone, the core components of the mouse skeletal system, depends on coordinated proliferation and differentiation of skeletogenic cells, including chondrocytes and osteoblasts. These cells differentiate from common progenitor cells originating in the mesoderm and neural crest. Multiple signaling pathways and transcription factors tightly regulate differentiation and proliferation of skeletal cells. In this chapter, we overview the process of mouse skeletal development and discuss major regulators of skeletal cells at each developmental stage.
Collapse
Affiliation(s)
- Tatsuya Kobayashi
- Massachusetts General Hospital, Harvard University, Boston, MA, USA.
| | | |
Collapse
|
37
|
Dasgupta K, Lessard S, Hann S, Fowler ME, Robling AG, Warman ML. Sensitive detection of Cre-mediated recombination using droplet digital PCR reveals Tg(BGLAP-Cre) and Tg(DMP1-Cre) are active in multiple non-skeletal tissues. Bone 2021; 142:115674. [PMID: 33031974 DOI: 10.1016/j.bone.2020.115674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
In humans, somatic activating mutations in PIK3CA are associated with skeletal overgrowth. In order to determine if activated PI3K signaling in bone cells causes overgrowth, we used Tg(BGLAP-Cre) and Tg(DMP1-Cre) mouse strains to somatically activate a disease-causing conditional Pik3ca allele (Pik3caH1047R) in osteoblasts and osteocytes. We observed Tg(BGLAP-Cre);Pik3caH1047R/+ offspring were born at the expected Mendelian frequency. However, these mice developed cutaneous lymphatic malformations and died before 7 weeks of age. In contrast, Tg(DMP1-Cre);Pik3caH1047R/+ offspring survived and had no cutaneous lymphatic malformations. Assuming that Cre-activity outside of the skeletal system accounted for the difference in phenotype between Tg(BGLAP-Cre);Pik3caH1047R/+ and Tg(DMP1-Cre);Pik3caH1047R/+ mice, we developed sensitive and specific droplet digital PCR (ddPCR) assays to search for and quantify rates of Tg(BGLAP-Cre)- and Tg(DMP1-Cre)-mediated recombination in non-skeletal tissues. We observed Tg(BGLAP-Cre)-mediated recombination in several tissues including skin, muscle, artery, and brain; two CNS locations, hippocampus and cerebellum, exhibited Cre-mediated recombination in >5% of cells. Tg(DMP1-Cre)-mediated recombination was also observed in muscle, artery, and brain. Although we cannot preclude that differences in phenotype between mice with Tg(BGLAP-Cre)- and Tg(DMP1-Cre)-mediated PIK3CA activation are due to Cre-recombination being induced at different stages of osteoblast differentiation, differences in recombination at non-skeletal sites are the more likely explanation. Since unanticipated sites of recombination can affect the interpretation of data from experiments involving conditional alleles, we recommend ddPCR as a good first step for assessing efficiency, leakiness, and off-targeting in experiments that employ Cre-mediated or Flp-mediated recombination.
Collapse
Affiliation(s)
- Krishnakali Dasgupta
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States of America; Department of Genetics, Harvard Medical School, Boston, MA, United States of America
| | - Samantha Lessard
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States of America
| | - Steven Hann
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States of America
| | - Megan E Fowler
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States of America
| | - Alexander G Robling
- Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Matthew L Warman
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States of America; Department of Genetics, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
38
|
Feng S, Bao L, Qiu G, Liao Z, Deng Z, Chen N, Chu Y, Luo Z, Jin Y, Li X, Yang Y, Zhao L. [Observation of dendrite osteocytes of mice at different developmental stages using Ploton silver staining and phalloidin staining]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1656-1661. [PMID: 33243734 DOI: 10.12122/j.issn.1673-4254.2020.11.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To assess the value of Ploton silver staining and phalloidin-iFlour 488 staining in observation of the morphology of osteocyte dendrites of mice at different developmental stages. METHODS The humerus and femurs were harvested from mice at 0 (P0), 5 (P5), 15 (P15), 21 (P21), 28 (P28), and 35 days (P35) after birth to prepare cryo-sections and paraffin sections. HE staining of P35 mouse femur sections served as a reference for observing osteocytes in the trabecular bone and cortical bone. The humeral sections at different developmental stages were stained with Ploton silver staining to observe the morphology of osteocytes and canaliculi, and the canalicular lengths in the cortical and trabecular bones of the humerus of the mice in each developmental stage were recorded. The cryo-sections of the humerus from P10 and P15 mice were stained with phalloidin iFlour-488 to observe the morphology of osteocytes and measurement of the length of osteocyte dendrites in the cortical bone. RESULTS In the trabecular bone of the humerus of P0-P15 mice, Ploton silver staining only visualized the outline of the osteocytes, and the morphology of the canaliculi was poorly defined. In P21 or older mice, Ploton silver staining revealed the morphology of the trabecular bone osteocytes and the canaliculi, which were neatly arranged and whose lengths increased significantly with age (P21 vs P28, P < 0.05; P21 vs P35, P < 0.05). In the humeral cortical bone of P15 mice, the morphology of the osteocytes and canalicular could be observed with Ploton silver staining, and the length of the regularly arranged canaliculi of the osteocytes increased significantly with age (P15 vs P21, P < 0.005; P15 vs P28, P < 0.0001; P15 vs P35, P < 0.0001). Phalloidin iFlour-488 staining was capable of visualizing the complete morphology of the osteocytes at P10 and P15; the osteocyte dendrites elongated progressively with age (P10 vs P15, P < 0.01) to form connections with the surrounding osteocytes. CONCLUSIONS Mouse osteocyte dendrites elongate progressively and their arrangement gradually becomes regular with age. Ploton silver staining can clearly visualize the morphology of the osteocytes and the canaliculi in adult mice but not in mice in early stages of development. Phalloidin iFlour-488 staining for labeling the cytoskeleton can be applied for mouse osteocytes at all developmental stages and allows morphological observation of mouse osteocytes in early developmental stages.
Collapse
Affiliation(s)
- Shuhao Feng
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Developmental Biology, Harvard School of Dental Medicine, Harvard University, Boston 02115, USA
| | - Liangxiao Bao
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Gengtao Qiu
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zheting Liao
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhonghao Deng
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Nachun Chen
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuhao Chu
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziheng Luo
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Jin
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyu Li
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard University, Boston 02115, USA
| | - Liang Zhao
- Department of Joint and Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
39
|
Yang YB, Goshtasebi A, van Lierop AH, Kalidasan D, Hitchcock CL, Prior JC. Effects of progesterone therapy on serum sclerostin levels in healthy menopausal women: a 3-month randomized, placebo-controlled clinical trial. Osteoporos Int 2020; 31:2243-2250. [PMID: 32564093 DOI: 10.1007/s00198-020-05505-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
UNLABELLED Sclerostin, a natural hormone made in bone, suppresses bone formation. Sclerostin is also decreased by estrogen. Progesterone, estrogen's menstrual partner, stimulates bone formation. It is unclear whether progesterone influences sclerostin. This study showed that progesterone did not change sclerostin using serum remaining from a randomized progesterone hot flush therapy trial. INTRODUCTION Progesterone and sclerostin are both endogenous hormones acting through osteoblast-origin cells and promote or suppress bone formation, respectively. Estradiol suppresses sclerostin, but progesterone, its menstrual cycle partner hormone, has unclear sclerostin relationships. We postulated that progesterone therapy would influence serum sclerostin levels. METHODS We obtained sclerostin levels for an ethics-approved post hoc analysis. Fasting sclerostin was measured in all remaining sera from a previous 12-week randomized controlled trial (RCT) of oral micronized progesterone (progesterone) for menopausal (> 1 year after last flow) vasomotor symptoms (VMS). Women in the RCT took 300 mg progesterone at bedtime or placebo (1:1) in a trial showing progesterone significantly decreased VMS. RESULTS Participants were healthy menopausal, primarily Caucasian (91.2%) community-dwelling women (± SD), 55.2 ± 4.6 years old with BMI 24.9 ± 2.9 kg/m2. The baseline sclerostin level in 60 women was 28.41 ± 10.47 pmol/L. Baseline sclerostin was not correlated with the run-in VMS score (r = 0.143, P = 0.294). Paired baseline and 12-week RCT data for 52 women showed serum sclerostin levels did not change related to experimental therapy (P = 0.504). Changes in final sclerostin values adjusted for baseline were progesterone (- 1.07 ± 7.96 pmol/L) and placebo (- 2.64 ± 8.70 pmol/L). In observational data (n = 60), baseline sclerostin levels correlated with the General Framingham Cardiovascular (CVD) Risk score (r = - 0.398, P = 0.003) and self-reported health by SF-36 quality of life instrument (QoL, r = - 0.331, P = 0.016). CONCLUSION Physiological oral micronized progesterone did not stimulate nor suppress serum sclerostin levels based on post hoc analysis of RCT data. Exploratory results, however, showed sclerostin negatively correlated with CVD risk and QoL. ClinicalTrials.gov #NCT0146469.
Collapse
Affiliation(s)
- Y B Yang
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - A Goshtasebi
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - A H van Lierop
- Center for Bone Quality, Leiden University Medical Center, Leiden, The Netherlands
| | - D Kalidasan
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - C L Hitchcock
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - J C Prior
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada.
- School of Population and Public Health, University of British Columbia, Vancouver, Canada.
- British Columbia Women's Health Research Institute, Vancouver, Canada.
- Department of Medicine, Division of Endocrinology and Metabolism, University of British Columbia, 2775 Laurel Street, Suite 4111, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
40
|
Eichholz KF, Woods I, Riffault M, Johnson GP, Corrigan M, Lowry MC, Shen N, Labour M, Wynne K, O'Driscoll L, Hoey DA. Human bone marrow stem/stromal cell osteogenesis is regulated via mechanically activated osteocyte-derived extracellular vesicles. Stem Cells Transl Med 2020; 9:1431-1447. [PMID: 32672416 PMCID: PMC7581449 DOI: 10.1002/sctm.19-0405] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/24/2020] [Accepted: 05/24/2020] [Indexed: 12/18/2022] Open
Abstract
Bone formation or regeneration requires the recruitment, proliferation, and osteogenic differentiation of stem/stromal progenitor cells. A potent stimulus driving this process is mechanical loading. Osteocytes are mechanosensitive cells that play fundamental roles in coordinating loading-induced bone formation via the secretion of paracrine factors. However, the exact mechanisms by which osteocytes relay mechanical signals to these progenitor cells are poorly understood. Therefore, this study aimed to demonstrate the potency of the mechanically stimulated osteocyte secretome in driving human bone marrow stem/stromal cell (hMSC) recruitment and differentiation, and characterize the secretome to identify potential factors regulating stem cell behavior and bone mechanobiology. We demonstrate that osteocytes subjected to fluid shear secrete a distinct collection of factors that significantly enhance hMSC recruitment and osteogenesis and demonstrate the key role of extracellular vesicles (EVs) in driving these effects. This demonstrates the pro-osteogenic potential of osteocyte-derived mechanically activated extracellular vesicles, which have great potential as a cell-free therapy to enhance bone regeneration and repair in diseases such as osteoporosis.
Collapse
Affiliation(s)
- Kian F. Eichholz
- Department of Mechanical, Aeronautical and Biomedical EngineeringMaterials and Surface Science Institute, University of LimerickLimerickIreland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Ian Woods
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Mathieu Riffault
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Gillian P. Johnson
- Department of Mechanical, Aeronautical and Biomedical EngineeringMaterials and Surface Science Institute, University of LimerickLimerickIreland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Michele Corrigan
- Department of Mechanical, Aeronautical and Biomedical EngineeringMaterials and Surface Science Institute, University of LimerickLimerickIreland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Michelle C. Lowry
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | - Nian Shen
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Marie‐Noelle Labour
- Department of Mechanical, Aeronautical and Biomedical EngineeringMaterials and Surface Science Institute, University of LimerickLimerickIreland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
| | - Kieran Wynne
- UCD Conway Institute of Biomolecular and Biomedical ResearchUniversity College DublinDublin 4Ireland
- Mass Spectrometry ResourceUniversity College DublinDublin 4Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | - David A. Hoey
- Department of Mechanical, Aeronautical and Biomedical EngineeringMaterials and Surface Science Institute, University of LimerickLimerickIreland
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences Institute, Trinity College DublinDublin 2Ireland
- Department of Mechanical and Manufacturing EngineeringSchool of Engineering, Trinity College DublinDublin 2Ireland
- Advanced Materials and Bioengineering Research CentreTrinity College Dublin & RCSIDublinIreland
| |
Collapse
|
41
|
Ruiz-Gaspà S, Guañabens N, Jurado S, Combalia A, Peris P, Monegal A, Parés A. Bilirubin and bile acids in osteocytes and bone tissue. Potential role in the cholestatic-induced osteoporosis. Liver Int 2020; 40:2767-2775. [PMID: 32749754 DOI: 10.1111/liv.14630] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Osteoporosis is a common complication in patients with primary biliary cholangitis. Both bilirubin and lithocholic acid (LCA) result in detrimental effects on osteoblastic cells, and ursodeoxycholic acid (UDCA) counteracts these outcomes. However, there is no information on the consequences of these retained substances of cholestasis and sera from cholestatic patients in osteocytes. METHODS The impact of bilirubin, LCA, UDCA and serum from jaundiced patients on viability, differentiation, mineralization and apoptosis has been assessed in MLO-Y4 and MLO-A5 osteocyte cell lines. Effects on gene expression were assessed in these cells and in human bone fragments. RESULTS Lithocholic acid 10 μmol/L and bilirubin 50 μmol/L decreased viability in MLO-Y4 and MLO-A5 cells (11% and 53% respectively; P ≤ .01). UDCA alone or combined with LCA or bilirubin increased cell viability. Jaundiced sera decreased cell viability (56%), an effect which was reverted by UDCA. Bilirubin decreased differentiation by 47% in MLO-Y4 (P ≤ .01) and mineralization (87%) after 21 days in MLO-A5 (P ≤ .03). Both bilirubin and LCA increased apoptosis in MLO-Y4, and UDCA diminished the apoptotic effect. Moreover, bilirubin down-regulated RUNX2 and up-regulated RANKL gene expression in bone tissue, MLO-Y4 and MLO-A5 cells, and LCA up-regulated RANKL expression in bone tissue. UDCA 100 μmol/L increased the gene expression of all these genes in bone tissue and MLO-Y4 cells and neutralized the decreased RUNX2 expression induced by bilirubin. CONCLUSION Bilirubin and LCA have damaging consequences in osteocytes by decreasing viability, differentiation and mineralization, increasing apoptosis and modifying gene expression, effects that are neutralized by UDCA.
Collapse
Affiliation(s)
- Silvia Ruiz-Gaspà
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Núria Guañabens
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Susana Jurado
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Andreu Combalia
- Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Pilar Peris
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Ana Monegal
- Metabolic Bone Diseases Unit, Department of Rheumatology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Albert Parés
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Liver Unit, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Cao W, Helder MN, Bravenboer N, Wu G, Jin J, Ten Bruggenkate CM, Klein-Nulend J, Schulten EAJM. Is There a Governing Role of Osteocytes in Bone Tissue Regeneration? Curr Osteoporos Rep 2020; 18:541-550. [PMID: 32676786 PMCID: PMC7532966 DOI: 10.1007/s11914-020-00610-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Bone regeneration plays an important role in contemporary clinical treatment. Bone tissue engineering should result in successful bone regeneration to restore congenital or acquired bone defects in the human skeleton. Osteocytes are thought to have a governing role in bone remodeling by regulating osteoclast and osteoblast activity, and thus bone loss and formation. In this review, we address the so far largely unknown role osteocytes may play in bone tissue regeneration. RECENT FINDINGS Osteocytes release biochemical signaling molecules involved in bone remodeling such as prostaglandins, nitric oxide, Wnts, and insulin-like growth factor-1 (IGF-1). Treatment of mesenchymal stem cells in bone tissue engineering with prostaglandins (e.g., PGE2, PGI2, PGF2α), nitric oxide, IGF-1, or Wnts (e.g., Wnt3a) improves osteogenesis. This review provides an overview of the functions of osteocytes in bone tissue, their interaction with other bone cells, and their role in bone remodeling. We postulate that osteocytes may have a pivotal role in bone regeneration as well, and consequently that the bone regeneration process may be improved effectively and rapidly if osteocytes are optimally used and stimulated.
Collapse
Affiliation(s)
- Wei Cao
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Marco N Helder
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Christiaan M Ten Bruggenkate
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Engelbert A J M Schulten
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Abstract
Skeleton formation and its proper functioning is possible thanks to specialized bone tissue
cells: bone forming osteoblasts, bone resorbing osteoclasts and osteocytes located in bone
cavities.
Gap junctions are transmembrane channels connecting neighboring cell. Thanks to gap junctions
it is possible for signals to be directly transmitted by cells. Gap junction type channels,
and more specifically the connexin proteins that build them, have a key impacton the bone
turnover process, and thus on both bone building and remodeling. A particularly important
connexin in bone tissue is connexin43 (Cx43), which is necessary in the proper course of the
bone formation process and in maintaining bone homeostasis.
The importance of the presence of Cx43 in bones is showed by skeletal defects in diseases
such as ODD syndrome and craniometaphyseal dysplasia caused by mutations in GJA1, the
gene encoding Cx43. The role of Cx43 in the differentiation of stem cells into bone cells,
anti-apoptotic action of bisphosphonates and bone responses to hormonal and mechanical
stimuli have also been demonstrated. In addition to connexin43, the presence of other connexins
such as connexin45, 46 and 37 was also noted in bone tissue.
Collapse
Affiliation(s)
- Krzysztof Łukowicz
- Zakład Biologii i Obrazowania Komórki, Instytut Zoologii i Badań Biomedycznych, Wydział Biologii, Uniwersytet Jagielloński w Krakowie
| | - Karolina Fijał
- Zakład Biologii i Obrazowania Komórki, Instytut Zoologii i Badań Biomedycznych, Wydział Biologii, Uniwersytet Jagielloński w Krakowie
| | - Aleksandra Nowak
- Zakład Biologii i Obrazowania Komórki, Instytut Zoologii i Badań Biomedycznych, Wydział Biologii, Uniwersytet Jagielloński w Krakowie
| | - Anna M. Osyczka
- Zakład Biologii i Obrazowania Komórki, Instytut Zoologii i Badań Biomedycznych, Wydział Biologii, Uniwersytet Jagielloński w Krakowie
| |
Collapse
|
44
|
Ahmadipour M, Mohammadi H, Pang AL, Arjmand M, Ayode Otitoju T, U. Okoye P, Rajitha B. A review: silicate ceramic-polymer composite scaffold for bone tissue engineering. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1817018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mohsen Ahmadipour
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Penang, Malaysia
| | - Hossein Mohammadi
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Penang, Malaysia
| | - Ai Ling Pang
- Faculty of Engineering, School of Chemical and Energy Engineering, UTM-MPRC Institute for Oil and Gas, Universiti Teknologi Malaysia, UTM Johor Bahru, Malaysia
| | - Mohammad Arjmand
- School of Engineering, University of British Columbia, Kelowna, BC, Canada
| | - Tunmise Ayode Otitoju
- School of Materials Science and Engineering, Shenyang University of Technology, Shenyang, China
| | - Patrick U. Okoye
- Laboratorio de Bioenergía, Instituto de Energías Renovables (IER-UNAM), Temixco, Morelos, México
| | - Beerelli Rajitha
- BVIRT Hyderabad College of Engineering for woman, Hyderabad, India
| |
Collapse
|
45
|
Expression of the type 1 lysophosphatidic acid receptor in osteoblastic cell lineage controls both bone mineralization and osteocyte specification. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158715. [DOI: 10.1016/j.bbalip.2020.158715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/03/2020] [Accepted: 04/10/2020] [Indexed: 11/23/2022]
|
46
|
Wei Y, Fu J, Wu W, Wu J. Comparative profiles of DNA methylation and differential gene expression in osteocytic areas from aged and young mice. Cell Biochem Funct 2020; 38:721-732. [PMID: 32526817 DOI: 10.1002/cbf.3539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/09/2020] [Accepted: 03/29/2020] [Indexed: 12/19/2022]
Abstract
Altered DNA methylation upon ageing may result in many age-related diseases such as osteoporosis. However, the changes in DNA methylation that occur in cortical bones, the major osteocytic areas, remain unknown. In our study, we extracted total DNA and RNA from the cortical bones of 6-month-old and 24-month-old mice and systematically analysed the differentially methylated regions (DMRs), differentially methylated promoters (DMPs) and differentially expressed genes (DEGs) between the mouse groups. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the DMR-related genes revealed that they were mainly associated with metabolic signalling pathways, including glycolysis, fatty acid and amino acid metabolism. Other genes with DMRs were related to signalling pathways that regulate the growth and development of cells, including the PI3K-AKT, Ras and Rap1 signalling pathways. The gene expression profiles indicated that the DEGs were mainly involved in metabolic pathways and the PI3K-AKT signalling pathway, and the profiles were verified through real-time quantitative PCR (RT-qPCR). Due to the pivotal roles of the affected genes in maintaining bone homeostasis, we suspect that these changes may be key factors in age-related bone loss, either together or individually. Our study may provide a novel perspective for understanding the osteocyte and its relationship with osteoporosis during ageing. SIGNIFICANCE OF THE STUDY: Our study identified age-related changes in gene expressions in osteocytic areas through whole-genome bisulfite sequencing (WGBS) and RNA-seq, providing new theoretical foundations for the targeted treatment of senile osteoporosis.
Collapse
Affiliation(s)
- Yu Wei
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jiayao Fu
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Wenjing Wu
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Junhua Wu
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
47
|
Jandl NM, von Kroge S, Stürznickel J, Baranowsky A, Stockhausen KE, Mushumba H, Beil FT, Püschel K, Amling M, Rolvien T. Large osteocyte lacunae in iliac crest infantile bone are not associated with impaired mineral distribution or signs of osteocytic osteolysis. Bone 2020; 135:115324. [PMID: 32198110 DOI: 10.1016/j.bone.2020.115324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 12/13/2022]
Abstract
The enlargement of osteocyte lacunae via osteocytic osteolysis was previously detected in situations of increased calcium demand (e.g., lactation, vitamin D deficiency). However, it is unclear whether similar processes occur also in the growing infantile skeleton and how this is linked to the mineral distribution within the bone matrix. Human iliac crest biopsies of 30 subjects (0-6 months, n = 14; 2-8 years, n = 6 and 18-25 years, n = 10) were acquired. Bone microarchitecture was assessed by micro-CT, while cellular bone histomorphometry was performed on undecalcified histological sections. Quantitative backscattered electron imaging (qBEI) was conducted to determine the bone mineral density distribution (BMDD) as well as osteocyte lacunar size and density. We additionally evaluated cathepsin K positive osteocytes using immunohistochemistry. Infantile bone was characterized by various signs of ongoing bone development such as higher bone (re)modeling, lower cortical and trabecular thickness compared to young adults. Importantly, a significantly higher osteocyte lacunar density and increased lacunar area were detected. Large osteocyte lacunae were associated with a more heterogeneous bone mineral density distribution of the trabecular bone matrix due to the presence of hypermineralized cartilage remnants, whereas the mean mineralization (i.e., CaMean) was not different in infantile bone. Absence of cathepsin K expression in osteocyte lacunae indicated nonexistent osteocytic osteolysis. Taken together, we demonstrated that the overall mineralization distribution in infantile bone is not altered compared to young adults besides high trabecular mineralization heterogeneity. Our study also provides important reference values for bone microstructure, BMDD and osteocyte characteristics in infants, children and young adults. Infantile bone displays large osteocyte lacunae indicating a developmental phenomenon rather than osteocytic osteolysis. Larger osteocytes may have superior mechanosensory abilities to enable bone adaption during growth.
Collapse
Affiliation(s)
- Nico Maximilian Jandl
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany
| | - Kilian Elia Stockhausen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany
| | - Herbert Mushumba
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| | - Frank Timo Beil
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Klaus Püschel
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, 22529 Hamburg, Germany
| | - Michael Amling
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529 Hamburg, Germany.
| |
Collapse
|
48
|
Zhang D, Li X, Pi C, Cai L, Liu Y, Du W, Yang W, Xie J. Osteoporosis-decreased extracellular matrix stiffness impairs connexin 43-mediated gap junction intercellular communication in osteocytes. Acta Biochim Biophys Sin (Shanghai) 2020; 52:517-526. [PMID: 32286624 DOI: 10.1093/abbs/gmaa025] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
Osteocytes are the main sensitive and responsive cells for mechanical stimuli in bone. The connexin family enables them to communicate with each other via forming functional gap junctions. However, how osteoporosis-impaired extracellular mechanical property modulates gap junction intercellular communication in osteocytes remains elusive. In this study, we established an ovariectomy (OVX)-induced osteoporosis mouse model in vivo and a polydimethylsiloxane (PDMS)-based cell culture substrate model in vitro to explore the influence of extracellular matrix (ECM) stiffness on cell-to-cell communication in osteocytes. Firstly, we established an OVX-induced osteoporosis mouse model by characterizing the changes in radiography, morphology and histochemistry of femurs. Our results showed that osteoporosis decreased the bone matrix stiffness together with the changes including the loss of osteocytes and the decrease of protein markers. Meanwhile, the dendritic process interconnection and channel-forming protein, Cx43, were reduced in osteoporosis mice. Next we mimicked ECM stiffness changes in vitro by using PDMS substrates at ratios 1:5 for normal stiffness and 1:45 for osteoporosis stiffness. Our results showed that the decreased ECM stiffness reduced the number of dendritic processes in a single cell and gap junctions between adjacent osteocytes. We further detected the decreased expression of Cx43, in the substrate with decreased stiffness. Finally, we found that gap junction-based intercellular communication was reduced in living osteocytes in the substrate with decreased stiffness. This study demonstrates the correlation between ECM mechanical property and cell-to-cell communication in osteocytes and might pave the way for further exploration of osteoporosis in terms of biomechanics.
Collapse
Affiliation(s)
- Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Wei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| |
Collapse
|
49
|
LaCombe JM, Roper RJ. Skeletal dynamics of Down syndrome: A developing perspective. Bone 2020; 133:115215. [PMID: 31887437 PMCID: PMC7044033 DOI: 10.1016/j.bone.2019.115215] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/05/2019] [Accepted: 12/24/2019] [Indexed: 01/14/2023]
Abstract
Individuals with Down syndrome (DS) display distinctive skeletal morphology compared to the general population, but disparate descriptions, methodologies, analyses, and populations sampled have led to diverging conclusions about this unique skeletal phenotype. As individuals with DS are living longer, they may be at a higher risk of aging disorders such as osteoporosis and increased fracture risk. Sexual dimorphism has been suggested between males and females with DS in which males, not females, experience an earlier decline in bone mineral density (BMD). Unfortunately, studies focusing on skeletal health related to Trisomy 21 (Ts21) are few in number and often too underpowered to answer questions about skeletal development, resultant osteoporosis, and sexual dimorphism, especially in stages of bone accrual. Further confounding the field are the varied methods of bone imaging, analysis, and data interpretation. This review takes a critical look at the current knowledge of DS skeletal phenotypes, both from human and mouse studies, and presents knowledge gaps that need to be addressed, differences in research methodologies and analyses that affect the interpretation of results, and proposes guidelines for overcoming obstacles to understand skeletal traits associated with DS. By examining our current knowledge of bone in individuals with Ts21, a trajectory for future studies may be established to provide meaningful solutions for understanding the development of and improving skeletal structures in individuals with and without DS.
Collapse
Affiliation(s)
- Jonathan M LaCombe
- Department of Biology, Indiana University-Purdue University Indianapolis, United States of America
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, United States of America.
| |
Collapse
|
50
|
Effect of fluoride on osteocyte-driven osteoclastic differentiation. Toxicology 2020; 436:152429. [PMID: 32156525 DOI: 10.1016/j.tox.2020.152429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 11/24/2022]
Abstract
Excessive systemic uptake of inorganic fluorides causes disturbances of bone homeostasis. The mechanism of skeletal fluorosis is still uncertain. This study aimed to study the effect of fluoride on osteocyte-driven osteoclastogenesis and probe into the role of PTH in this process. IDG-SW3 cells seeded in collagen-coated constructs were developed into osteocyte-like cells through induction of mineral agents. Then, osteocyte-like cells were exposed to fluoride in the presence or absence of parathyroid hormone (PTH). Cell viability and their capacity to produce receptor activator of nuclear factor kappa-B ligand (RANKL), osteoprotegerin (OPG) and sclerostin (SOST) were detected by MTT and Western blot assays, respectively. Finally, a transwell coculture system using osteocyte-like cells seeded in the low compartment, and osteoclast precursors added in the inserts was developed to observe the osteocyte-driven osteoclasogenesis response to fluoride with or without PTH, and the expression of molecules involved in this mechanism were measure by real time RT-PCR. Results showed that osteocytes withstood a toxic dose of fluoride, and yet PTH administration significantly reduced osteocytes viability. PTH amplified the effect of fluoride on the expression of osteoclastogenesis-related molecules in osteocyte, but did not enlarged the stimulating effect of fluoride on osteoclastogenesis drove by osteocyte coculture. Gene expression levels of TRAP, RANK, JNK and NFAtc1 significantly increased in fluoride affected osteoclast precursor cocultured with osteocyte-like cells. The impact of fluoride on osteocyte-driven osteoclast differentiation was stronger than that of PTH. In conclusion, osteocyte played a pivotal role on the mechanism underlying fluoride-affected osteoclastogenesis in which RANK-JNK-NFATc1 signaling pathway was involved, and PTH had a significant impact in this process.
Collapse
|