51
|
Aw Yong KM, Horst E, Neale D, Royzenblat S, Lahann J, Greineder C, Weivoda M, Mehta G, Keller ET. A Bioreactor for 3D In Vitro Modeling of the Mechanical Stimulation of Osteocytes. Front Bioeng Biotechnol 2022; 10:797542. [PMID: 35402411 PMCID: PMC8990130 DOI: 10.3389/fbioe.2022.797542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
The bone is a mechanosensitive organ that is also a common metastatic site for prostate cancer. However, the mechanism by which the tumor interacts with the bone microenvironment to further promote disease progression remains to be fully understood. This is largely due to a lack of physiological yet user-friendly models that limit our ability to perform in-depth mechanistic studies. Here, we report a tunable bioreactor which facilitates the 3D culture of the osteocyte cell line, MLO-Y4, in a hydroxyapatite/tricalcium phosphate (HA/TCP) scaffold under constant fluidic shear stress and tunable hydrostatic pressure within physiological parameters. Increasing hydrostatic pressure was sufficient to induce a change in the expression of several bone remodeling genes such as Dmp1, Rankl, and Runx2. Furthermore, increased hydrostatic pressure induced the osteocytes to promote the differentiation of the murine macrophage cell line RAW264.7 toward osteoclast-like cells. These results demonstrate that the bioreactor recapitulates the mechanotransduction response of osteocytes to pressure including the measurement of their functional ability in a 3D environment. In conclusion, the bioreactor would be useful for exploring the mechanisms of osteocytes in bone health and disease.
Collapse
Affiliation(s)
- Koh Meng Aw Yong
- Department of Urology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Eric Horst
- Department of Material Science and Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Dylan Neale
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Sonya Royzenblat
- Biosciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Joerg Lahann
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biosciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Colin Greineder
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Megan Weivoda
- Biosciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Geeta Mehta
- Department of Material Science and Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Evan T. Keller
- Department of Urology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
- Biosciences Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
52
|
Fielding C, García-García A, Korn C, Gadomski S, Fang Z, Reguera JL, Pérez-Simón JA, Göttgens B, Méndez-Ferrer S. Cholinergic signals preserve haematopoietic stem cell quiescence during regenerative haematopoiesis. Nat Commun 2022; 13:543. [PMID: 35087060 PMCID: PMC8795384 DOI: 10.1038/s41467-022-28175-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/12/2022] [Indexed: 12/15/2022] Open
Abstract
The sympathetic nervous system has been evolutionary selected to respond to stress and activates haematopoietic stem cells via noradrenergic signals. However, the pathways preserving haematopoietic stem cell quiescence and maintenance under proliferative stress remain largely unknown. Here we found that cholinergic signals preserve haematopoietic stem cell quiescence in bone-associated (endosteal) bone marrow niches. Bone marrow cholinergic neural signals increase during stress haematopoiesis and are amplified through cholinergic osteoprogenitors. Lack of cholinergic innervation impairs balanced responses to chemotherapy or irradiation and reduces haematopoietic stem cell quiescence and self-renewal. Cholinergic signals activate α7 nicotinic receptor in bone marrow mesenchymal stromal cells leading to increased CXCL12 expression and haematopoietic stem cell quiescence. Consequently, nicotine exposure increases endosteal haematopoietic stem cell quiescence in vivo and impairs hematopoietic regeneration after haematopoietic stem cell transplantation in mice. In humans, smoking history is associated with delayed normalisation of platelet counts after allogeneic haematopoietic stem cell transplantation. These results suggest that cholinergic signals preserve stem cell quiescence under proliferative stress.
Collapse
Affiliation(s)
- Claire Fielding
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Andrés García-García
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Claudia Korn
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Stephen Gadomski
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
- NIH-Oxford-Cambridge Scholars Program in partnership with Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zijian Fang
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Juan L Reguera
- Department of Hematology, University Hospital Virgen del Rocio, 41013, Sevilla, Spain
| | - José A Pérez-Simón
- NIH-Oxford-Cambridge Scholars Program in partnership with Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Hematology, University Hospital Virgen del Rocio, 41013, Sevilla, Spain
| | - Berthold Göttgens
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK.
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK.
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK.
- Instituto de Biomedicina de Sevilla (IBiS/CSIC), Universidad de Sevilla, 41013, Seville, Spain.
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, 41009, Seville, Spain.
| |
Collapse
|
53
|
Brady RT, O’Brien FJ, Hoey DA. The Impact of the Extracellular Matrix Environment on Sost Expression by the MLO-Y4 Osteocyte Cell Line. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9010035. [PMID: 35049744 PMCID: PMC8772728 DOI: 10.3390/bioengineering9010035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/27/2022]
Abstract
Bone is a dynamic organ that can adapt its structure to meet the demands of its biochemical and biophysical environment. Osteocytes form a sensory network throughout the tissue and orchestrate tissue adaptation via the release of soluble factors such as a sclerostin. Osteocyte physiology has traditionally been challenging to investigate due to the uniquely mineralized extracellular matrix (ECM) of bone leading to the development of osteocyte cell lines. Importantly, the most widely researched and utilized osteocyte cell line: the MLO-Y4, is limited by its inability to express sclerostin (Sost gene) in typical in-vitro culture. We theorised that culture in an environment closer to the in vivo osteocyte environment could impact on Sost expression. Therefore, this study investigated the role of composition and dimensionality in directing Sost expression in MLO-Y4 cells using collagen-based ECM analogues. A significant outcome of this study is that MLO-Y4 cells, when cultured on a hydroxyapatite (HA)-containing two-dimensional (2D) film analogue, expressed Sost. Moreover, three-dimensional (3D) culture within HA-containing collagen scaffolds significantly enhanced Sost expression, demonstrating the impact of ECM composition and dimensionality on MLO-Y4 behaviour. Importantly, in this bone mimetic ECM environment, Sost expression was found to be comparable to physiological levels. Lastly, MLO-Y4 cells cultured in these novel conditions responded accordingly to fluid flow stimulation with a decrease in expression. This study therefore presents a novel culture system for the MLO-Y4 osteocyte cell line, ensuring the expression of an important osteocyte specific gene, Sost, overcoming a major limitation of this model.
Collapse
Affiliation(s)
- Robert T. Brady
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (R.T.B.); (F.J.O.)
- Trinity Centre for Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, D02 PN40 Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (R.T.B.); (F.J.O.)
- Trinity Centre for Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, D02 PN40 Dublin, Ireland
| | - David A. Hoey
- Trinity Centre for Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, D02 PN40 Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Correspondence:
| |
Collapse
|
54
|
Abstract
Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Teresita Bellido
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas,3Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
55
|
Day RN, Day KH, Pavalko FM. Direct visualization by FRET-FLIM of a putative mechanosome complex involving Src, Pyk2 and MBD2 in living MLO-Y4 cells. PLoS One 2021; 16:e0261660. [PMID: 34941939 PMCID: PMC8699642 DOI: 10.1371/journal.pone.0261660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023] Open
Abstract
Earlier, we proposed the “mechanosome” concept as a testable model for understanding how mechanical stimuli detected by cell surface adhesion molecules are transmitted to modulate gene expression inside cells. Here, for the first time we document a putative mechanosome involving Src, Pyk2 and MBD2 in MLO-Y4 osteocytes with high spatial resolution using FRET-FLIM. Src-Pyk2 complexes were concentrated at the periphery of focal adhesions and the peri-nuclear region. Pyk2-MBD2 complexes were located primarily in the nucleus and peri-nuclear region. Lifetime measurements indicated that Src and MBD2 did not interact directly. Finally, mechanical stimulation by fluid flow induced apparent accumulation of Src-Pyk2 protein complexes in the peri-nuclear/nuclear region, consistent with the proposed behavior of a mechanosome in response to a mechanical stimulus.
Collapse
Affiliation(s)
- Richard N. Day
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kathleen H. Day
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Fredrick M. Pavalko
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
56
|
Effect of High Static Magnetic Fields on Biological Activities and Iron Metabolism in MLO-Y4 Osteocyte-like Cells. Cells 2021; 10:cells10123519. [PMID: 34944025 PMCID: PMC8700585 DOI: 10.3390/cells10123519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/29/2022] Open
Abstract
There are numerous studies that investigate the effects of static magnetic fields (SMFs) on osteoblasts and osteoclasts. However, although osteocytes are the most abundant cell type in bone tissue, there are few studies on the biological effects of osteocytes under magnetic fields. Iron is a necessary microelement that is involved in numerous life activities in cells. Studies have shown that high static magnetic fields (HiSMF) can regulate cellular iron metabolism. To illustrate the effect of HiSMF on activities of osteocytes, and whether iron is involved in this process, HiSMF of 16 tesla (T) was used, and the changes in cellular morphology, cytoskeleton, function-related protein expression, secretion of various cytokines, and iron metabolism in osteocytes under HiSMF were studied. In addition, the biological effects of HiSMF combined with iron preparation and iron chelator on osteocytes were also investigated. The results showed that HiSMF promoted cellular viability, decreased apoptosis, increased the fractal dimension of the cytoskeleton, altered the secretion of cytokines, and increased iron levels in osteocytes. Moreover, it was found that the biological effects of osteocytes under HiSMF are attenuated or enhanced by treatment with a certain concentration of iron. These data suggest that HiSMF-regulated cellular iron metabolism may be involved in altering the biological effects of osteocytes under HiSMF exposure.
Collapse
|
57
|
Liu Y, Duan M, Guo D, Kan S, Zhang L, Aili M, Zhang D, Du W, Xie J. PDGF-AA promotes cell-to-cell communication in osteocytes through PI3K/Akt signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1640-1649. [PMID: 34586354 DOI: 10.1093/abbs/gmab136] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/24/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Osteocytes are the main sensitive cells in bone remodeling due to their potent functional cell processes from the mineralized bone matrix to the bone surface and the bone marrow. Neighboring osteocytes communicate with each other by these cell processes to achieve molecular exchange through gap junction channels. Platelet-derived growth factor-AA (PDGF-AA) has been reported to enhance bone tissue remodeling by promoting cell proliferation, migration, and autocrine secretion in osteoid cell linage. However, the effect of PDGF-AA on intercellular communication between osteocytes is still unclear. In the present study, we elucidated that PDGF-AA could enhance the formation of dendritic processes of osteocytes and the gap junctional intercellular communication by promoting the expression of connexin43 (Cx43). This modulation process was mainly dependent on the activation of phosphorylation of Akt protein by phosphatidylinositol 3-kinase (PI3K)/Akt (also known as protein kinase B, PKB) signaling. Inhibition of PI3K/Akt signaling decreased the Cx43 expression induced by PDGF-AA. These results establish a bridge between PDGF-AA and cell-cell communication in osteocytes, which could help us understand the molecular exchange between bone cells and fracture healing.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Daimo Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Shiyi Kan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Li Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Munire Aili
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Demao Zhang
- 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
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
- Department of Cariology and Endodontics, 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
|
58
|
Yu H, Zhang J, Liu X, Li Y. microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway. Bone Joint Res 2021; 10:744-758. [PMID: 34847690 PMCID: PMC8712601 DOI: 10.1302/2046-3758.1012.bjr-2020-0275.r2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aims Exosomes derived from bone marrow mesenchymal stem cells (BMSCs) have been reported to be a promising cellular therapeutic approach for various human diseases. The current study aimed to investigate the mechanism of BMSC-derived exosomes carrying microRNA (miR)-136-5p in fracture healing. Methods A mouse fracture model was initially established by surgical means. Exosomes were isolated from BMSCs from mice. The endocytosis of the mouse osteoblast MC3T3-E1 cell line was analyzed. CCK-8 and disodium phenyl phosphate microplate methods were employed to detect cell proliferation and alkaline phosphatase (ALP) activity, respectively. The binding of miR-136-5p to low-density lipoprotein receptor related protein 4 (LRP4) was analyzed by dual luciferase reporter gene assay. HE staining, tartrate-resistant acid phosphatase (TRAP) staining, and immunohistochemistry were performed to evaluate the healing of the bone tissue ends, the positive number of osteoclasts, and the positive expression of β-catenin protein, respectively. Results miR-136-5p promoted fracture healing and osteoblast proliferation and differentiation. BMSC-derived exosomes exhibited an enriched miR-136-5p level, and were internalized by MC3T3-E1 cells. LRP4 was identified as a downstream target gene of miR-136-5p. Moreover, miR-136-5p or exosomes isolated from BMSCs (BMSC-Exos) containing miR-136-5p activated the Wnt/β-catenin pathway through the inhibition of LRP4 expression. Furthermore, BMSC-derived exosomes carrying miR-136-5p promoted osteoblast proliferation and differentiation, thereby promoting fracture healing. Conclusion BMSC-derived exosomes carrying miR-136-5p inhibited LRP4 and activated the Wnt/β-catenin pathway, thus facilitating fracture healing. Cite this article: Bone Joint Res 2021;10(12):744–758.
Collapse
Affiliation(s)
- Haichi Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Jun Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Xiaoning Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Yingzhi Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
59
|
Mechanical loading activates the YAP/TAZ pathway and chemokine expression in the MLO-Y4 osteocyte-like cell line. J Transl Med 2021; 101:1597-1604. [PMID: 34521992 DOI: 10.1038/s41374-021-00668-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022] Open
Abstract
Osteocytes are mechanosensitive cells that control bone remodeling in response to mechanical loading. To date, specific signaling pathways modulated by mechanical loading in osteocytes are not well understood. Yes associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), the main effectors of the Hippo pathway, are reported to play a role in mechanotransduction and during osteoblastogenesis. Here, we hypothesized that YAP/TAZ signaling mediates osteocyte mechanosensing to target genes of the bone remodeling process. We aimed to investigate the contribution of YAP/TAZ in modulating the gene expression in an osteocyte-like cell line MLO-Y4. We developed a 3D osteocyte compression culture model from an MLO-Y4 osteocyte cell line embedded in concentrated collagen hydrogel. 3D-mechanical loading led to the increased expression of mechanosensitive genes and a subset of chemokines, including M-csf, Cxcl1, Cxcl2, Cxcl3, Cxcl9, and Cxcl10. The transcription regulators YAP and TAZ translocated to the nucleus and upregulated their target genes and proteins. RNAseq analysis revealed that YAP/TAZ knockdown mediated the regulation of several genes including osteocyte dendrite formation. Use of YAP/TAZ knockdown partially blunted the increase in M-csf and Cxcl3 levels in response to MLO-Y4 compression. These findings demonstrate that YAP/TAZ signaling is required for osteocyte-like cell mechano-transduction, regulates the gene expression profiles and controls chemokine expression.
Collapse
|
60
|
Wang XY, Ma TL, Chen KN, Pang ZY, Wang H, Huang JM, Qi GB, Wang CZ, Jiang ZX, Gong LJ, Wang Z, Jiang C, Yan ZQ. Accumulation of LDL/ox-LDL in the necrotic region participates in osteonecrosis of the femoral head: a pathological and in vitro study. Lipids Health Dis 2021; 20:167. [PMID: 34823555 PMCID: PMC8620162 DOI: 10.1186/s12944-021-01601-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a common but intractable disease that appears to involve lipid metabolic disorders. Although numerous studies have demonstrated that high blood levels of low-density lipoprotein (LDL) are closely associated with ONFH, there is limited evidence to explain the pathological role of LDL. Pathological and in vitro studies were performed to investigate the role of disordered metabolism of LDL and oxidized LDL (ox-LDL) in the femoral head in the pathology of ONFH. METHODS Nineteen femoral head specimens from patients with ONFH were obtained for immunohistochemistry analysis. Murine long-bone osteocyte Y4 cells were used to study the effects of LDL/ox-LDL on cell viability, apoptosis, and metabolism process of LDL/ox-LDL in osteocytes in normoxic and hypoxic environments. RESULTS In the pathological specimens, marked accumulation of LDL/ox-LDL was observed in osteocytes/lacunae of necrotic regions compared with healthy regions. In vitro studies showed that ox-LDL, rather than LDL, reduced the viability and enhanced apoptosis of osteocytes. Pathological sections indicated that the accumulation of ox-LDL was significantly associated with impaired blood supply. Exposure to a hypoxic environment appeared to be a key factor leading to LDL/ox-LDL accumulation by enhancing internalisation and oxidation of LDL in osteocytes. CONCLUSIONS The accumulation of LDL/ox-LDL in the necrotic region may contribute to the pathology of ONFH. These findings could provide new insights into the prevention and treatment of ONFH.
Collapse
Affiliation(s)
- Xin-Yuan Wang
- Department of Orthopaedics, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Tian-Le Ma
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Kang-Ning Chen
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhi-Ying Pang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Hao Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jun-Ming Huang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Guo-Bin Qi
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chen-Zhong Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zeng-Xin Jiang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Lin-Jing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhe Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chang Jiang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Zuo-Qin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
61
|
Generation of two multipotent mesenchymal progenitor cell lines capable of osteogenic, mature osteocyte, adipogenic, and chondrogenic differentiation. Sci Rep 2021; 11:22593. [PMID: 34799645 PMCID: PMC8605002 DOI: 10.1038/s41598-021-02060-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 11/09/2021] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal progenitors differentiate into several tissues including bone, cartilage, and adipose. Targeting these cells in vivo is challenging, making mesenchymal progenitor cell lines valuable tools to study tissue development. Mesenchymal stem cells (MSCs) can be isolated from humans and animals; however, obtaining homogenous, responsive cells in a reproducible fashion is challenging. As such, we developed two mesenchymal progenitor cell (MPC) lines, MPC1 and MPC2, generated from bone marrow of male C57BL/6 mice. These cells were immortalized using the temperature sensitive large T-antigen, allowing for thermal control of proliferation and differentiation. Both MPC1 and MPC2 cells are capable of osteogenic, adipogenic, and chondrogenic differentiation. Under osteogenic conditions, both lines formed mineralized nodules, and stained for alizarin red and alkaline phosphatase, while expressing osteogenic genes including Sost, Fgf23, and Dmp1. Sost and Dmp1 mRNA levels were drastically reduced with addition of parathyroid hormone, thus recapitulating in vivo responses. MPC cells secreted intact (iFGF23) and C-terminal (cFGF23) forms of the endocrine hormone FGF23, which was upregulated by 1,25 dihydroxy vitamin D (1,25D). Both lines also rapidly entered the adipogenic lineage, expressing adipose markers after 4 days in adipogenic media. MPC cells were also capable of chondrogenic differentiation, displaying increased expression of cartilaginous genes including aggrecan, Sox9, and Comp. With the ability to differentiate into multiple mesenchymal lineages and mimic in vivo responses of key regulatory genes/proteins, MPC cells are a valuable model to study factors that regulate mesenchymal lineage allocation as well as the mechanisms that dictate transcription, protein modification, and secretion of these factors.
Collapse
|
62
|
Sibilia V, Bottai D, Maggi R, Pagani F, Chiaramonte R, Giannandrea D, Citro V, Platonova N, Casati L. Sex Steroid Regulation of Oxidative Stress in Bone Cells: An In Vitro Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182212168. [PMID: 34831936 PMCID: PMC8621144 DOI: 10.3390/ijerph182212168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023]
Abstract
Environmental stimuli, including sex hormones and oxidative stress (OS), affect bone balance, modifying the epigenetic profiles of key osteogenic genes. Nonetheless, the interplay between sex steroids, epigenome and OS has yet be fully elucidated. This paper aims to study in vitro the role of sex steroids in OS-induced alteration in bone cells’ homeostasis, and to assess the possible contribution of epigenetic modifications. Toward this purpose, osteoblast (MC3T3-E1) and osteocyte (MLOY-4) cell lines were exposed to two different sources of free oxygen radicals, i.e., tert-butyl hydroperoxide and dexamethasone, and the protective effect of pre-treatment with androgens and estrogens was evaluated. In particular, we analyzed parameters that reflect bone cell homeostasis such as cell viability, cell migration, transcriptomic profile, transcriptional activity, and epigenetic signature. Our findings indicate that estrogens and androgens counteract OS effects. Using partially overlapping strategies, they reduce OS outcomes regarding cell viability, cell migration, the transcriptomic profile of gene families involved in bone remodeling, and epigenetic profile, i.e., H3K4me3 level. Additionally, we demonstrated that the protective effect of steroids against OS on bone homeostasis is partially mediated by the Akt pathway. Overall, these results suggest that the hormonal milieu may influence the mechanisms of age-related bone disease.
Collapse
Affiliation(s)
- Valeria Sibilia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milano, Italy; (V.S.); (F.P.)
| | - Daniele Bottai
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
| | - Roberto Maggi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milano, Italy;
| | - Francesca Pagani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20129 Milano, Italy; (V.S.); (F.P.)
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
| | - Domenica Giannandrea
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
| | - Valentina Citro
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
| | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
| | - Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy; (D.B.); (R.C.); (D.G.); (V.C.); (N.P.)
- Correspondence:
| |
Collapse
|
63
|
Matsuzaka T, Matsugaki A, Nakano T. Control of osteoblast arrangement by osteocyte mechanoresponse through prostaglandin E2 signaling under oscillatory fluid flow stimuli. Biomaterials 2021; 279:121203. [PMID: 34717197 DOI: 10.1016/j.biomaterials.2021.121203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 10/07/2021] [Accepted: 10/20/2021] [Indexed: 01/04/2023]
Abstract
Anisotropic collagen/apatite microstructure is a prominent determinant of bone tissue functionalization; in particular, bone matrix modulates its anisotropic microstructure depending on the surrounding mechanical condition. Although mechanotransduction in bones is governed by osteocyte function, the precise mechanisms linking mechanical stimuli and anisotropic formation of collagen/apatite microstructure are poorly understood. Here we developed a novel anisotropic mechano-coculture system which enables the understanding of the biological mechanisms regulating the oriented bone matrix formation, which is constructed by aligned osteoblasts. The developed model provides bone-mimetic coculture platform that enables simultaneous control of mechanical condition and osteoblast-osteocyte communication with an anisotropic culture scaffold. The engineered coculture device helps in understanding the relationship between osteocyte mechanoresponses and osteoblast arrangement, which is a significant contributor to anisotropic organization of bone tissue. Our study showed that osteocyte responses to oscillatory flow stimuli regulated osteoblast arrangement through soluble molecular interactions. Importantly, we found that prostaglandin E2 is a novel determinant for oriented collagen/apatite organization of bone matrix, through controlling osteoblast arrangement.
Collapse
Affiliation(s)
- Tadaaki Matsuzaka
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Aira Matsugaki
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
64
|
Abstract
MicroRNAs, identified in the early 1990s, are believed to regulate approximately 30% of the human genome. The role of microRNA in bone cells was first reported in 2007 in a manuscript showing that microRNA-223 is essential for osteoclast differentiation in vitro, and a few studies reported a role of microRNAs in osteoblasts the same year. The first report of microRNA actions in osteocytes was published in 2010, in which it was demonstrated that the microRNA cluster 23a~27a~24-2 regulates osteocyte differentiation. Since then, few studies have described the role of these 18-25-nucleotide non-coding RNAs on osteocyte biology, reporting osteocytes both as producers and as targets of the actions of microRNAs. We review here the current knowledge on the effects of microRNAs on osteocyte biology.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, USA; Indiana Center for Musculoskeletal Health, USA.
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
65
|
Kurihara M, Mukudai Y, Watanabe H, Asakura M, Abe Y, Houri A, Chikuda J, Shimane T, Shirota T. Autophagy prevents osteocyte cell death under hypoxic conditions. Cells Tissues Organs 2021; 210:326-338. [PMID: 34412050 DOI: 10.1159/000519086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Mai Kurihara
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Yoshiki Mukudai
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Hitoshi Watanabe
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Mariko Asakura
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Yuzo Abe
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Asami Houri
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Junichiro Chikuda
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Toshikazu Shimane
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
66
|
Eichholz KF, Federici A, Riffault M, Woods I, Mahon OR, O’Driscoll L, Hoey DA. Extracellular Vesicle Functionalized Melt Electrowritten Scaffolds for Bone Tissue Engineering. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Kian F. Eichholz
- Department of Mechanical, Aeronautical and Biomedical Engineering Materials and Surface Science Institute University of Limerick Limerick Ireland
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Angelica Federici
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Mathieu Riffault
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Ian Woods
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Olwyn R. Mahon
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
| | - Lorraine O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Trinity St. James's Cancer Institute Trinity College Dublin Dublin Ireland
| | - David A. Hoey
- Department of Mechanical, Aeronautical and Biomedical Engineering Materials and Surface Science Institute University of Limerick Limerick Ireland
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute Trinity College Dublin Dublin Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering Trinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre Trinity College Dublin & RCSI Dublin Ireland
| |
Collapse
|
67
|
Fournier R, Harrison RE. Methods for studying MLO-Y4 osteocytes in collagen-hydroxyapatite scaffolds in the rotary cell culture system. Connect Tissue Res 2021; 62:436-453. [PMID: 32375524 DOI: 10.1080/03008207.2020.1764548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: The rotary cell culture system (RCCS) is a common clinorotation device for cell culture. It is also used as a low-shear suspension culture bioreactor to form functionalized 3D tissue constructs and to model microgravity. We sought to develop a 3D scaffold composed of type I collagen and hydroxyapatite (collagen-HA) to characterize MLO-Y4 osteocytes following suspension culture or clinorotation.Materials and Methods: MLO-Y4 cells were embedded in collagen-HA. The scaffold was formed into droplets for suspension culture or wall-adhered to the RCCS for clinorotation. AFM, rheometry, immunofluorescence and qRT-PCR were employed to measure the scaffold stiffness, cell viability and gene expression of cells in collagen-HA scaffolds. Dendritic cells were visualized and quantified and gene expression after suspension culture and clinorotation was compared to static controls.Results: The optimized scaffold for the RCCS consisted of collagen with 6 mg/mL HA which had a stiffness of < 1 kPa. MLO-Y4 cell viability was higher in collagen-HA scaffolds, compared to scaffolds without HA. Collagen-HA scaffolds induced higher osteocyte-specific gene expression compared to cells cultured on 2D plastic. Cells in the scaffold downregulated DMP1, E11, IL-6, and RANKL, and had fewer dendritic cells following suspension culture whereas clinorotation downregulated DMP1 and E11 genes, compared to static controls.Conclusions: Suspension culture for 3 days in collagen-HA stimulates growth of osteocytes but may also desensitize them to mechanical cues. Clinorotation for 3 days in collagen-HA does not stimulate proliferation or expression of mechanosensitive genes, indicating that it may be an effective mechanical unloading environment.
Collapse
Affiliation(s)
- Roxanne Fournier
- Department of Cell & Systems Biology and the Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Rene E Harrison
- Department of Cell & Systems Biology and the Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
68
|
Miyama A, Ebina K, Hirao M, Okamura G, Etani Y, Takami K, Goshima A, Miura T, Oyama S, Kanamoto T, Yoshikawa H, Nakata K. Effects of iguratimod on glucocorticoid-induced disorder of bone metabolism in vitro. J Bone Miner Metab 2021; 39:639-648. [PMID: 33564917 DOI: 10.1007/s00774-021-01206-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Glucocorticoids are widely used to treat various diseases including rheumatoid arthritis (RA); however, one of the most frequent and severe adverse effects is glucocorticoid-induced osteoporosis (GIOP). Iguratimod (IGU) is a novel conventional synthetic disease-modifying anti-rheumatic drug developed in Japan. The aim of this study is to investigate the effects of IGU on glucocorticoid-induced disorder of bone metabolism in vitro. MATERIALS AND METHODS In osteoclastogenesis of mouse bone marrow-derived cells, tartrate-resistant acid phosphatase staining, resorption pit assay, western blotting, real-time polymerase chain reaction (PCR), and mRNA sequencing were performed. In osteoblastogenesis of MC3T3-E1 cells, alkaline phosphatase (ALP) staining and activity, alizarin red staining, and mRNA sequencing were performed, and real-time PCR and western blotting were conducted in MC3T3-E1 cells and murine osteocyte-like cell line MLO-Y4 cells. RESULTS IGU significantly suppressed a dexamethasone-induced increase in osteoclasts, differentiation, and bone resorption activity by inhibition of the receptor activator of the nuclear factor kappa-B (RANK)/tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6)/nuclear factor kappa-B (NFκB)-p52 pathway. In MC3T3-E1 cells, IGU significantly upregulated dexamethasone-induced downregulation of ALP activity, bone mineralization, and osteoblast-related gene and protein expression. In MLO-Y4 cells, IGU significantly upregulated dexamethasone-induced downregulation of the gene expression of ALP and osteocalcin, and also downregulated receptor activator of NFκB ligand (RANKL)/osteoprotegerin gene expression ratio without dexamethasone. CONCLUSION These results suggest that IGU may improve glucocorticoid-induced disorder of bone metabolism and may exhibit positive effects against GIOP associated with RA.
Collapse
Affiliation(s)
- Akira Miyama
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Makoto Hirao
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Gensuke Okamura
- Department of Orthopaedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasone-cho, Kita-ku, Sakai, 591-8025, Japan
| | - Yuki Etani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kenji Takami
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Taihei Miura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shohei Oyama
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takashi Kanamoto
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka, 560-8565, Japan
| | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
69
|
Multiomic analysis of stretched osteocytes reveals processes and signalling linked to bone regeneration and cancer. NPJ Regen Med 2021; 6:32. [PMID: 34099736 PMCID: PMC8184808 DOI: 10.1038/s41536-021-00141-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 05/10/2021] [Indexed: 11/08/2022] Open
Abstract
Exercise is a non-pharmacological intervention that can enhance bone regeneration and improve the management of bone conditions like osteoporosis or metastatic bone cancer. Therefore, it is gaining increasing importance in an emerging area of regenerative medicine-regenerative rehabilitation (RR). Osteocytes are mechanosensitive and secretory bone cells that orchestrate bone anabolism and hence postulated to be an attractive target of regenerative exercise interventions. However, the human osteocyte signalling pathways and processes evoked upon exercise remain to be fully identified. Making use of a computer-controlled bioreactor that mimics exercise and the latest omics approaches, RNA sequencing (RNA-seq) and tandem liquid chromatography-mass spectrometry (LC-MS), we mapped the transcriptome and secretome of mechanically stretched human osteocytic cells. We discovered that a single bout of cyclic stretch activated network processes and signalling pathways likely to modulate bone regeneration and cancer. Furthermore, a comparison between the transcriptome and secretome of stretched human and mouse osteocytic cells revealed dissimilar results, despite both species sharing evolutionarily conserved signalling pathways. These findings suggest that osteocytes can be targeted by exercise-driven RR protocols aiming to modulate bone regeneration or metastatic bone cancer.
Collapse
|
70
|
Qin L, Fu X, Ma J, Lin M, Zhang P, Wang Y, Yan Q, Tao C, Liu W, Tang B, Chen D, Bai X, Cao H, Xiao G. Kindlin-2 mediates mechanotransduction in bone by regulating expression of Sclerostin in osteocytes. Commun Biol 2021; 4:402. [PMID: 33767359 PMCID: PMC7994671 DOI: 10.1038/s42003-021-01950-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Osteocytes act as mechanosensors in bone; however, the underlying mechanism remains poorly understood. Here we report that deleting Kindlin-2 in osteocytes causes severe osteopenia and mechanical property defects in weight-bearing long bones, but not in non-weight-bearing calvariae. Kindlin-2 loss in osteocytes impairs skeletal responses to mechanical stimulation in long bones. Control and cKO mice display similar bone loss induced by unloading. However, unlike control mice, cKO mice fail to restore lost bone after reloading. Osteocyte Kindlin-2 deletion impairs focal adhesion (FA) formation, cytoskeleton organization and cell orientation in vitro and in bone. Fluid shear stress dose-dependently increases Kindlin-2 expression and decreases that of Sclerostin by downregulating Smad2/3 in osteocytes; this latter response is abolished by Kindlin-2 ablation. Kindlin-2-deficient osteocytes express abundant Sclerostin, contributing to bone loss in cKO mice. Collectively, we demonstrate an indispensable novel role of Kindlin-2 in maintaining skeletal responses to mechanical stimulation by inhibiting Sclerostin expression during osteocyte mechanotransduction.
Collapse
Affiliation(s)
- Lei Qin
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Xuekun Fu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Jing Ma
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Manxia Lin
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Peijun Zhang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Yishu Wang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Wen Liu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
71
|
Jablonská E, Horkavcová D, Rohanová D, Brauer DS. A review of in vitro cell culture testing methods for bioactive glasses and other biomaterials for hard tissue regeneration. J Mater Chem B 2021; 8:10941-10953. [PMID: 33169773 DOI: 10.1039/d0tb01493a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bioactive glasses are used to regenerate bone by a mechanism which involves surface degradation, the release of ions such as calcium, soluble silica and phosphate and the precipitation of a biomimetic apatite surface layer on the glass. One major area of bioactive glass research is the incorporation of therapeutically active ions to broaden the application range of these materials. When developing such new compositions, in vitro cell culture studies are a key part of their characterisation. However, parameters of cell culture studies vary widely, and depending on the intended use of bioactive glass compositions, different layouts, cell types and assays need to be used. The aim of this publication is to provide materials scientists, particularly those new to cell culture studies, with a tool for selecting the most appropriate assays to give insight into the properties of interest.
Collapse
Affiliation(s)
- Eva Jablonská
- Laboratory of Molecular Biology and Virology, Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague 6, Czech Republic.
| | - Diana Horkavcová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Dana Rohanová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Delia S Brauer
- Otto Schott Institute of Materials Research, Faculty of Chemistry and Earth Sciences, Friedrich Schiller University Jena, Fraunhoferstr. 6, 07743 Jena, Germany.
| |
Collapse
|
72
|
Chang B, Liu X. Osteon: Structure, Turnover, and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:261-278. [PMID: 33487116 DOI: 10.1089/ten.teb.2020.0322] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone is composed of dense and solid cortical bone and honeycomb-like trabecular bone. Although cortical bone provides the majority of mechanical strength for a bone, there are few studies focusing on cortical bone repair or regeneration. Osteons (the Haversian system) form structural and functional units of cortical bone. In recent years, emerging evidences have shown that the osteon structure (including osteocytes, lamellae, lacunocanalicular network, and Haversian canals) plays critical roles in bone mechanics and turnover. Therefore, reconstruction of the osteon structure is crucial for cortical bone regeneration. This article provides a systematic summary of recent advances in osteons, including the structure, function, turnover, and regenerative strategies. First, the hierarchical structure of osteons is illustrated and the critical functions of osteons in bone dynamics are introduced. Next, the modeling and remodeling processes of osteons at a cellular level and the turnover of osteons in response to mechanical loading and aging are emphasized. Furthermore, several bioengineering approaches that were recently developed to recapitulate the osteon structure are highlighted. Impact statement This review provides a comprehensive summary of recent advances in osteons, especially the roles in bone formation, remodeling, and regeneration. Besides introducing the hierarchical structure and critical functions of osteons, we elucidate the modeling and remodeling of osteons at a cellular level. Specifically, we highlight the bioengineering approaches that were recently developed to mimic the hierarchical structure of osteons. We expect that this review will provide informative insights and attract increasing attentions in orthopedic community, shedding light on cortical bone regeneration in the future.
Collapse
Affiliation(s)
- Bei Chang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| |
Collapse
|
73
|
Riquelme MA, Gu S, Hua R, Jiang JX. Mechanotransduction via the coordinated actions of integrins, PI3K signaling and Connexin hemichannels. Bone Res 2021; 9:8. [PMID: 33531460 PMCID: PMC7854719 DOI: 10.1038/s41413-020-00126-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/25/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanical loading opens connexin 43 (Cx43) hemichannels (HCs), leading to the release of bone anabolic molecules, such as prostaglandins, from mechanosensitive osteocytes, which is essential for bone formation and remodeling. However, the mechanotransduction mechanism that activates HCs remains elusive. Here, we report a unique pathway by which mechanical signals are effectively transferred between integrin molecules located in different regions of the cell, resulting in HC activation. Both integrin α5 and αV were activated upon mechanical stimulation via either fluid dropping or flow shear stress (FSS). Inhibition of integrin αV activation or ablation of integrin α5 prevented HC opening on the cell body when dendrites were mechanically stimulated, suggesting mechanical transmission from the dendritic integrin αV to α5 in the cell body during HC activation. In addition, HC function was compromised in vivo, as determined by utilizing an antibody blocking αV activation and α5-deficient osteocyte-specific knockout mice. Furthermore, inhibition of integrin αV activation, but not that of α5, attenuated activation of the phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway upon mechanical loading, and the inhibition of PI3K/AKT activation blocked integrin α5 activation and HC opening. Moreover, HC opening was blocked only by an anti-integrin αV antibody at low but not high FSS levels, suggesting that dendritic αV is a more sensitive mechanosensor than α5 for activating HCs. Together, these results reveal a new molecular mechanism of mechanotransduction involving the coordinated actions of integrins and PI3K/AKT in osteocytic dendritic processes and cell bodies that leads to HC opening and the release of key bone anabolic factors.
Collapse
Affiliation(s)
- Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
74
|
Negrescu AM, Necula MG, Gebaur A, Golgovici F, Nica C, Curti F, Iovu H, Costache M, Cimpean A. In Vitro Macrophage Immunomodulation by Poly(ε-caprolactone) Based-Coated AZ31 Mg Alloy. Int J Mol Sci 2021; 22:ijms22020909. [PMID: 33477539 PMCID: PMC7831122 DOI: 10.3390/ijms22020909] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Due to its excellent bone-like mechanical properties and non-toxicity, magnesium (Mg) and its alloys have attracted great interest as biomaterials for orthopaedic applications. However, their fast degradation rate in physiological environments leads to an acute inflammatory response, restricting their use as biodegradable metallic implants. Endowing Mg-based biomaterials with immunomodulatory properties can help trigger a desired immune response capable of supporting a favorable healing process. In this study, electrospun poly(ε-caprolactone) (PCL) fibers loaded with coumarin (CM) and/or zinc oxide nanoparticles (ZnO) were used to coat the commercial AZ31 Mg alloy as single and combined formulas, and their effects on the macrophage inflammatory response and osteoclastogenic process were investigated by indirect contact studies. Likewise, the capacity of the analyzed samples to generate reactive oxygen species (ROS) has been investigated. The data obtained by attenuated total reflection Fourier-transform infrared (FTIR-ATR) and X-ray photoelectron spectroscopy (XPS) analyses indicate that AZ31 alloy was perfectly coated with the PCL fibers loaded with CM and ZnO, which had an important influence on tuning the release of the active ingredient. Furthermore, in terms of degradation in phosphate-buffered saline (PBS) solution, the PCL-ZnO- and secondary PCL-CM-ZnO-coated samples exhibited the best corrosion behaviour. The in vitro results showed the PCL-CM-ZnO and, to a lower extent, PCL-ZnO coated sample exhibited the best behaviour in terms of inflammatory response and receptor activator of nuclear factor kappa-B ligand (RANKL)-mediated differentiation of RAW 264.7 macrophages into osteoclasts. Altogether, the results obtained suggest that the coating of Mg alloys with fibrous PCL containing CM and/or ZnO can constitute a feasible strategy for biomedical applications.
Collapse
Affiliation(s)
- Andreea-Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Madalina-Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Adi Gebaur
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Florentina Golgovici
- Department of General Chemistry, Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania;
| | - Cristina Nica
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Filis Curti
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Horia Iovu
- Advance Polymer Materials Group, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Gh. Polizu 17, 011061 Bucharest, Romania; (A.G.); (F.C.); (H.I.)
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (A.-M.N.); (M.-G.N.); (C.N.); (M.C.)
- Correspondence: ; Tel.: +40-21-318-1575 (ext. 106)
| |
Collapse
|
75
|
Palmieri M, Kim HN, Gomez-Acevedo H, Que X, Tsimikas S, Jilka RL, Manolagas SC, Witztum JL, Ambrogini E. A Neutralizing Antibody Targeting Oxidized Phospholipids Promotes Bone Anabolism in Chow-Fed Young Adult Mice. J Bone Miner Res 2021; 36:170-185. [PMID: 32990984 PMCID: PMC7855899 DOI: 10.1002/jbmr.4173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/14/2020] [Accepted: 08/23/2020] [Indexed: 12/16/2022]
Abstract
Oxidized phospholipids containing phosphocholine (OxPL) are pro-inflammatory lipid peroxidation products that bind to scavenger receptors (SRs), such as Scarb1, and toll-like receptors (TLRs). Excessive OxPL, as found in oxidized low-density lipoprotein (OxLDL), overwhelm these defense mechanisms and become pathogenic in atherosclerosis, nonalcoholic steatohepatitis (NASH), and osteoporosis. We previously reported that the innate IgM natural antibody E06 binds to OxPL and neutralizes their deleterious effects; expression of the single-chain (scFv) form of the antigen-binding domain of E06 (E06-scFv) as a transgene increases trabecular bone in male mice. We show herein that E06-scFv increases trabecular and cortical bone in female and male mice by increasing bone formation and decreasing osteoblast apoptosis in vivo. Homozygous E06-scFv mice have higher bone mass than hemizygous, showing a dose effect of the transgene. To investigate how OxPL restrain bone formation under physiologic conditions, we measured the levels of SRs and TLRs that bind OxPL. We found that osteoblastic cells primarily express Scarb1. Moreover, OxLDL-induced apoptosis and reduced differentiation were prevented in bone marrow-derived or calvaria-derived osteoblasts from Scarb1 knockout mice. Because Scarb1-deficient mice are reported to have high bone mass, our results suggest that E06 may promote bone anabolism in healthy young mice, at least in part, by neutralizing OxPL, which in turn promote Scarb1-mediated apoptosis of osteoblasts or osteoblast precursors. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)..
Collapse
Affiliation(s)
- Michela Palmieri
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Ha-Neui Kim
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Horacio Gomez-Acevedo
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xuchu Que
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
76
|
Shimasaki M, Ueda S, Ichiseki T, Hirata H, Kawahara N, Ueda Y. Resistance of bone marrow mesenchymal stem cells in a stressed environment - Comparison with osteocyte cells. Int J Med Sci 2021; 18:1375-1381. [PMID: 33628093 PMCID: PMC7893571 DOI: 10.7150/ijms.52104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/17/2020] [Indexed: 11/05/2022] Open
Abstract
Introduction: Recently, the efficacy of mesenchymal stem cells (MSCs) mediated by their tissue repair and anti-inflammatory actions in the prevention and therapy of various disorders has been reported. In this research, our attention was focused specifically on the prevention and therapy of glucocorticoid-induced osteonecrosis. We investigated the stress resistance of MSC against glucocorticoid administration and hypoxic stress, which are factors known to induce osteocytic cell death. Materials and Methods: Mouse bone cells (MLO-Y4) and bone-marrow derived mouse MSCs were exposed to dexamethasone (Dex), hypoxia of 1% oxygen or both in vitro. Mitochondrial membrane potentials were estimated by mitochondria labeling with a cell-permeant probe (Mito tracker red); expression of these apoptosis-inducing molecules, oxidative stress marker (8-hydroxy-2'-deoxyguanosine), caspase-3, -9, and two apoptosis-inhibiting molecules, energy-producing ATP synthase (ATP5A) and X-linked inhibitor of apoptosis protein (XIAP), were analyzed by both immunofluorescence and western blot. Results: With exposure to either dexamethasone or hypoxia, MLO-Y4 showed reduced mitochondrial membrane potential, ATP5A and upregulation of 8-OHdG, cleaved caspases and XIAP. Those changes were significantly enhanced by treatment with dexamethasone plus hypoxia. In MSCs, however, mitochondrial membrane potentials were preserved, while no significant changes in the pro-apoptosis or anti-apoptosis molecules analyzed were found even with exposure to both dexamethasone and hypoxia. No such effects induced by treatment with dexamethasone, hypoxia, or both were demonstrated in MSCs at all. Discussion: In osteocyte cells subjected to the double stresses of glucocorticoid administration and a hypoxic environment osteocytic cell death was mediated via mitochondria. In contrast, MSC subjected to the same stressors showed preservation of mitochondrial function and reduced oxidative stress. Accordingly, even under conditions sufficiently stressful to cause the osteocytic cell death in vivo, it was thought that the function of MSC could be preserved, suggesting that in the case of osteonecrosis preventative and therapeutic strategies incorporating their intraosseous implantation may be promising.
Collapse
Affiliation(s)
- Miyako Shimasaki
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| | - Shusuke Ueda
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Toru Ichiseki
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Hiroaki Hirata
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Norio Kawahara
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Yoshimichi Ueda
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| |
Collapse
|
77
|
Abstract
Osteocytes are an ancient cell, appearing in fossilized skeletal remains of early fish and dinosaurs. Despite its relative high abundance, even in the context of nonskeletal cells, the osteocyte is perhaps among the least studied cells in all of vertebrate biology. Osteocytes are cells embedded in bone, able to modify their surrounding extracellular matrix via specialized molecular remodeling mechanisms that are independent of the bone forming osteoblasts and bone-resorbing osteoclasts. Osteocytes communicate with osteoclasts and osteoblasts via distinct signaling molecules that include the RankL/OPG axis and the Sost/Dkk1/Wnt axis, among others. Osteocytes also extend their influence beyond the local bone environment by functioning as an endocrine cell that controls phosphate reabsorption in the kidney, insulin secretion in the pancreas, and skeletal muscle function. These cells are also finely tuned sensors of mechanical stimulation to coordinate with effector cells to adjust bone mass, size, and shape to conform to mechanical demands.
Collapse
Affiliation(s)
- Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| |
Collapse
|
78
|
Wang Z, Weng Y, Ishihara Y, Odagaki N, Ei Hsu Hlaing E, Izawa T, Okamura H, Kamioka H. Loading history changes the morphology and compressive force-induced expression of receptor activator of nuclear factor kappa B ligand/osteoprotegerin in MLO-Y4 osteocytes. PeerJ 2020; 8:e10244. [PMID: 33240612 PMCID: PMC7659647 DOI: 10.7717/peerj.10244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In this study, we investigated the effect of the mechanical loading history on the expression of receptor activator of nuclear factor kappa B ligand (RANKL) and osteoprotegerin (OPG) in MLO-Y4 osteocyte-like cells. METHODS Three hours after MLO-Y4 osteocytes were seeded, a continuous compressive force (CCF) of 31 dynes/cm2 with or without additional CCF (32 dynes/cm2) was loaded onto the osteocytes. After 36 h, the additional CCF (loading history) was removed for a recovery period of 10 h. The expression of RANKL, OPG, RANKL/OPG ratio, cell numbers, viability and morphology were time-dependently examined at 0, 3, 6 and 10 h. Then, the same additional CCF was applied again for 1 h to all osteocytes with or without the gap junction inhibitor to examine the expression of RANKL, OPG, the RANKL/OPG ratio and other genes that essential to characterize the phenotype of MLO-Y4 cells. Fluorescence recovery after photobleaching technique was also applied to test the differences of gap-junctional intercellular communications (GJIC) among MLO-Y4 cells. RESULTS The expression of RANKL and OPG by MLO-Y4 osteocytes without a loading history was dramatically decreased and increased, respectively, in response to the 1-h loading of additional weight. However, the expression of RANKL, OPG and the RANKL/OPG ratio were maintained at the same level as in the control group in the MLO-Y4 osteocytes with a loading history but without gap junction inhibitor treatment. Treatment of loading history significantly changed the capacity of GJIC and protein expression of connexin 43 (Cx43) but not the mRNA expression of Cx43. No significant difference was observed in the cell number or viability between the MLO-Y4 osteocyte-like cells with and without a loading history or among different time checkpoints during the recovery period. The cell morphology showed significant changes and was correlated with the expression of OPG, Gja1 and Dmp1 during the recovery period. CONCLUSION Our findings indicated that the compressive force-induced changes in the RANKL/OPG expression could be habituated within at least 11 h by 36-h CCF exposure. GJIC and cell morphology may play roles in response to loading history in MLO-Y4 osteocyte-like cells.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yao Weng
- Department of Oral Morphology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshihito Ishihara
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Naoya Odagaki
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ei Ei Hsu Hlaing
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takashi Izawa
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
79
|
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
|
80
|
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
|
81
|
Abstract
The aim of this review was to compile a list of tools currently available to study bone cells and in particular osteocytes. As the interest (and importance) in osteocyte biology has greatly expanded over the past decade, new tools and techniques have become available to study these elusive cells, RECENT FINDINGS: Osteocytes are the main orchestrators of bone remodeling. They control both osteoblasts and osteoclast activities via cell-to cell communication or through secreted factors. Osteocytes are also the mechanosensors of the bone and they orchestrate skeletal adaptation to loads. Recent discoveries have greatly expanded our knowledge and understanding of these cells and new models are now available to further uncover the functions of osteocytes. Novel osteocytic cell lines, primary cultures, and 3D scaffolds are now available to investigators to further unravel the functions and roles of these cells.
Collapse
Affiliation(s)
- Paola Divieti Pajevic
- Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, 700 Albany Street, W201E, Boston, MA, 02118, USA.
| |
Collapse
|
82
|
Isolation of Murine and Human Osteocytes. Methods Mol Biol 2020. [PMID: 32979194 DOI: 10.1007/978-1-0716-0989-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Osteocytes are thought to be the mechanosensors of bone by sensing mechanical loads imposed upon the bone and transmitting these signals to the other bone cells to initiate bone modeling and remodeling. The location of osteocytes deep within bone is ideal for their function. However, this location makes the study of osteocytes in vivo technically difficult. There are several methods for obtaining and culturing primary osteocytes for in vitro experiments and ex vivo observation. In this chapter, several proven methods are discussed including the isolation of avian osteocytes from chicks and osteocytes from calvaria and long bones of young mice. A detailed protocol for the isolation of osteocytes from hypermineralized bone of mature and aged animals is provided. In addition, a modified version of this protocol that can be used to isolate osteocytes from human trabecular bone is described.
Collapse
|
83
|
Loy TL, Vehlow D, Kauschke V, Müller M, Heiss C, Lips KS. Effects of BDNF and PEC Nanoparticles on Osteocytes. Molecules 2020; 25:molecules25184151. [PMID: 32927875 PMCID: PMC7570603 DOI: 10.3390/molecules25184151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Bone substitute materials loaded with mediators that stimulate fracture healing are demanded in the clinical treatment in trauma surgery and orthopedics. Brain-derived neurotrophic factor (BDNF) enhances the proliferation and differentiation of mesenchymal stem cells into osteoblast. To load the implants with BDNF, a drug delivery system that allows the release of BDNF under spatiotemporal control would improve functionality. Polyelectrolyte complex nanoparticles (PECNP) have been reported as a suitable drug delivery system. The suitability of PECNP in contact with osteocytes as the main cell type of bone is not known so far. Thus, we aimed to verify that BDNF and PECNP loaded with BDNF (PECNP+BDNF) as well as pure PECNP have no negative effects on osteocytes in vitro. Therefore, the murine osteocyte cell line MLO-Y4 was treated with BDNF and PECNP+BDNF. The effects on proliferation were analyzed by the BrdU test (n = 5). The results demonstrated a significant increase in proliferation 24 h after BDNF application, whereas PECNP+BDNF did not lead to significant changes. Thus, we conclude that BDNF is an appropriate mediator to stimulate osteocytes. Since the addition of PECNP did not affect the viability of osteocytes, we conclude that PECNP are a suitable drug delivery system for bone implants.
Collapse
Affiliation(s)
- Thomas Leonhard Loy
- Experimental Trauma Surgery, Justus-Liebig-University, 35392 Giessen, Germany; (T.L.L.); (V.K.); (C.H.)
| | - David Vehlow
- Department Functional Colloidal Materials, Leibniz Institute of Polymer Research, 01069 Dresden, Germany; (D.V.); (M.M.)
| | - Vivien Kauschke
- Experimental Trauma Surgery, Justus-Liebig-University, 35392 Giessen, Germany; (T.L.L.); (V.K.); (C.H.)
| | - Martin Müller
- Department Functional Colloidal Materials, Leibniz Institute of Polymer Research, 01069 Dresden, Germany; (D.V.); (M.M.)
| | - Christian Heiss
- Experimental Trauma Surgery, Justus-Liebig-University, 35392 Giessen, Germany; (T.L.L.); (V.K.); (C.H.)
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital of Giessen-Marburg GmbH, Campus Giessen, 35392 Giessen, Germany
| | - Katrin Susanne Lips
- Experimental Trauma Surgery, Justus-Liebig-University, 35392 Giessen, Germany; (T.L.L.); (V.K.); (C.H.)
- Correspondence: ; Tel.: +49-641-99-30580
| |
Collapse
|
84
|
Wang Y, Han B, Ding J, Qiu C, Wang W. Endoplasmic reticulum stress mediates osteocyte death under oxygen-glucose deprivation in vitro. Acta Histochem 2020; 122:151577. [PMID: 32778239 DOI: 10.1016/j.acthis.2020.151577] [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: 04/01/2020] [Revised: 06/06/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
As a vascularized organ, bone is known to be susceptible to ischemia. Ischemic osteonecrosis or skeletal unloading lead to ischemia in bone microenvironment that causes osteocytes to suffer hypoxia and nutrition deprivation. OBJECTIVE To explore the effects of Oxygen-glucose deprivation (OGD) on osteocytes and the potential mechanism. METHODS OGD model was established in cultured MLO-Y4 cell. Cell damage, intracellular oxidative stress and cell apoptosis were detected at different OGD times (0, 2, 4, 8, 12, 24 h), and the changes in endoplasmic reticulum (ER) stress-related indicators were observed. Furthermore, cells were treated with 4-phenylbutyrate sodium (4-PBA) to inhibit ER stress, and cell damage and oxidative stress level were detected. RESULTS The cell viability under OGD exhibited a significantly reduced in a time-dependent manner, and the level of intracellular reactive oxygen species (ROS) were increased, cell apoptosis and ER stress was induced. Inhibition of ER stress can reduce cell death and intracellular ROS levels. CONCLUSION Our study demonstrated that ER stress regulates OGD-induced apoptotic cell death in MLO-Y4 cells via intracellular ROS.
Collapse
|
85
|
Uruç V, Salimov F, Mahir Kapl H. Protective Effect of Hypericum perforatum Extract on Methotrexate-Induced Osteotoxicity via Reducing Oxidative Stress and MAPK Activity. INT J PHARMACOL 2020. [DOI: 10.3923/ijp.2020.430.436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
86
|
Wang XY, Gong LJ, Huang JM, Jiang C, Yan ZQ. Pinocembrin alleviates glucocorticoid-induced apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway in osteocytes. Eur J Pharmacol 2020; 880:173212. [PMID: 32470335 DOI: 10.1016/j.ejphar.2020.173212] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Glucocorticoids are widely used in clinical practice, but are associated with potentially severe side effects like glucocorticoid-induced osteoporosis (GIOP) and glucocorticoid-associated osteonecrosis of the femoral head (GA-ONFH). Glucocorticoid-induced osteocyte apoptosis plays critical roles in the pathological processes of both GIOP and GA-ONFH. Pinocembrin is a natural flavonoid that may exert protective effects on osteocytes. The present study investigated the effects of pinocembrin on glucocorticoid-induced apoptosis of murine long bone osteocyte Y4 (MLO-Y4) cells and sought to elucidate the underlying molecular mechanism. We found that pinocembrin attenuated glucocorticoid-induced cell viability injury and apoptosis of MLO-Y4 cells. Moreover, pinocembrin increased Beclin-1 and LC3B-II level, but decreased p62 expression, suggesting that pinocembrin activates autophagy in glucocorticoid-treated MLO-Y4 cells. The protective effects of pinocembrin on glucocorticoid-induced apoptosis of MLO-Y4 cells were mimicked by a known stimulator of autophagy but prevented by a known inhibitor of autophagy. Pinocembrin also suppressed the PI3K/Akt/mTOR signaling pathway, which regulates cell autophagy, in glucocorticoid-treated MLO-Y4 cells. In conclusion, the results indicate that pinocembrin alleviates glucocorticoid-induced osteocyte apoptosis by activating autophagy via suppressing the PI3K/Akt/mTOR pathway. Pinocembrin may represent a potential natural agent for preventing and treating GIOP and GA-ONFH.
Collapse
Affiliation(s)
- Xin-Yuan Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Lin-Jing Gong
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jun-Ming Huang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Chang Jiang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zuo-Qin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
87
|
Schröder M, Riksen EA, He J, Skallerud BH, Møller ME, Lian AM, Syversen U, Reseland JE. Vitamin K2 Modulates Vitamin D-Induced Mechanical Properties of Human 3D Bone Spheroids In Vitro. JBMR Plus 2020; 4:e10394. [PMID: 32995695 PMCID: PMC7507351 DOI: 10.1002/jbm4.10394] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Rotational culture promotes primary human osteoblasts (hOBs) to form three-dimensional (3D) multicellular spheroids with bone tissue-like structure without any scaffolding material. Cell-based bone models enable us to investigate the effect of different agents on the mechanical strength of bone. Given that low dietary intake of both vitamin D and K is negatively associated with fracture risk, we aimed to assess the effect of these vitamins in this system. Osteospheres of hOBs were generated with menaquinone-4 (MK-4; 10μM) and 25-hydroxyvitamin D3 [25(OH)D3; 0.01μM], alone and in combination, or without vitamins. The mechanical properties were tested by nanoindentation using a flat-punch compression method, and the mineralized extracellular bone matrix was characterized by microscopy. The in vitro response of hOBs to MK-4 and 25(OH)D3 was further evaluated in two-dimensional (2D) cultures and in the 3D bone constructs applying gene expression analysis and multiplex immunoassays. Mechanical testing revealed that 25(OH)D3 induced a stiffer and MK-4 a softer or more flexible osteosphere compared with control. Combined vitamin conditions induced the same flexibility as MK-4 alone. Enhanced levels of periostin (p < 0.001) and altered distribution of collagen type I (COL-1) were found in osteospheres supplemented with MK-4. In contrast, 25(OH)D3 reduced COL-1, both at the mRNA and protein levels, increased alkaline phosphatase, and stimulated mineral deposition in the osteospheres. With the two vitamins in combination, enhanced gene expression of periostin and COL-1 was seen, as well as extended osteoid formation into the central region and increased mineral deposition all over the area. Moreover, we observed enhanced levels of osteocalcin in 2D and osteopontin in 3D cultures exposed to 25(OH)D3 alone and combined with MK-4. In conclusion, the two vitamins seem to affect bone mechanical properties differently: vitamin D enhancing stiffness and K2 conveying flexibility to bone. These effects may translate to increased fracture resistance in vivo. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Maria Schröder
- Department of Biomaterials University of Oslo Oslo Norway
| | | | - Jianying He
- Department of Structural Engineering, Faculty of Engineering Norwegian University of Science and Technology (NTNU) Trondheim Norway
| | - Bjørn Helge Skallerud
- Department of Structural Engineering, Faculty of Engineering Norwegian University of Science and Technology (NTNU) Trondheim Norway
| | | | - Aina-Mari Lian
- Oral Research Laboratory, Institute for Clinical Dentistry University of Oslo Oslo Norway
| | - Unni Syversen
- Oral Research Laboratory, Institute for Clinical Dentistry University of Oslo Oslo Norway.,Department of Clinical and Molecular Medicine NTNU Trondheim Norway.,Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital Trondheim University Hospital Trondheim Norway
| | | |
Collapse
|
88
|
Oliveira MC, Pieters BCH, Guimarães PB, Duffles LF, Heredia JE, Silveira ALM, Oliveira ACC, Teixeira MM, Ferreira AVM, Silva TA, van de Loo FAJ, Macari S. Bovine Milk Extracellular Vesicles Are Osteoprotective by Increasing Osteocyte Numbers and Targeting RANKL/OPG System in Experimental Models of Bone Loss. Front Bioeng Biotechnol 2020; 8:891. [PMID: 32850743 PMCID: PMC7411003 DOI: 10.3389/fbioe.2020.00891] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Studying effects of milk components on bone may have a clinical impact as milk is highly associated with bone maintenance, and clinical studies provided controversial associations with dairy consumption. We aimed to evaluate the impact of milk extracellular vesicles (mEVs) on the dynamics of bone loss in mice. MEVs are nanoparticles containing proteins, mRNA and microRNA, and were supplemented into the drinking water of mice, either receiving diet-induced obesity or ovariectomy (OVX). Mice receiving mEVs were protected from the bone loss caused by diet-induced obesity. In a more severe model of bone loss, OVX, higher osteoclast numbers in the femur were found, which were lowered by mEV treatment. Additionally, the osteoclastogenic potential of bone marrow-derived precursor cells was lowered in mEV-treated mice. The reduced stiffness in the femur of OVX mice was consequently reversed by mEV treatment, accompanied by improvement in the bone microarchitecture. In general, the RANKL/OPG ratio increased systemically and locally in both models and was rescued by mEV treatment. The number of osteocytes, as primary regulators of the RANKL/OPG system, raised in the femur of the OVX mEVs-treated group compared to OVX non-treated mice. Also, the osteocyte cell line treated with mEVs demonstrated a lowered RANKL/OPG ratio. Thus, mEVs showed systemic and local osteoprotective properties in two mouse models of bone loss reflected in reduced osteoclast presence. Data reveal mEV potential in bone modulation, acting via osteocyte enhancement and RANKL/OPG regulation. We suggest that mEVs could be a therapeutic candidate for the treatment of bone loss.
Collapse
Affiliation(s)
- Marina C Oliveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bartijn C H Pieters
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Polianna B Guimarães
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Letícia F Duffles
- Department of Oral Surgery and Pathology, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Joyce E Heredia
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana L M Silveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Amanda C C Oliveira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adaliene V M Ferreira
- Laboratory of Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tarcilia A Silva
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Oral Surgery and Pathology, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fons A J van de Loo
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Soraia Macari
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Restorative Dentistry, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
89
|
Anesi A, Malavasi G, Chiarini L, Salvatori R, Lusvardi G. Cell Proliferation to Evaluate Preliminarily the Presence of Enduring Self-Regenerative Antioxidant Activity in Cerium Doped Bioactive Glasses. MATERIALS 2020; 13:ma13102297. [PMID: 32429291 PMCID: PMC7288167 DOI: 10.3390/ma13102297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
(1) Background: a cell evaluation focused to verify the self-regenerative antioxidant activity is performed on cerium doped bioactive glasses. (2) Methods: the glasses based on 45S5 Bioglass®, are doped with 1.2 mol%, 3.6 mol% and 5.3 mol% of CeO2 and possess a polyhedral shape (~500 µm2). Glasses with this composition inhibit oxidative stress by mimicking catalase enzyme (CAT) and superoxide dismutase (SOD) activities; moreover, our previous cytocompatibility tests (neutral red (NR), 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Bromo-2-deoxyUridine (BrdU)) reveal that the presence of cerium promotes the absorption and vitality of the cells. The same cytocompatibility tests were performed and repeated, in two different periods (named first and second use), separated from each other by four months. (3) Results: in the first and second use, NR tests indicate that the presence of cerium promotes once again cell uptake and viability, especially after 72 h. A decrease in cell proliferation it is observed after MTT and BrdU tests only in the second use. These findings are supported by statistically significant results (4) Conclusions: these glasses show enhanced proliferation, both in the short and in the long term, and for the first time such large dimensions are studied for this kind of study. A future prospective is the implantation of these bioactive glasses as bone substitute in animal models.
Collapse
Affiliation(s)
- Alexandre Anesi
- SMECHIMAI, Università di Modena e Reggio Emilia, Largo del Pozzo 71, 41125 Modena, Italy; (A.A.); (L.C.); (R.S.)
| | - Gianluca Malavasi
- DSCG, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy;
| | - Luigi Chiarini
- SMECHIMAI, Università di Modena e Reggio Emilia, Largo del Pozzo 71, 41125 Modena, Italy; (A.A.); (L.C.); (R.S.)
| | - Roberta Salvatori
- SMECHIMAI, Università di Modena e Reggio Emilia, Largo del Pozzo 71, 41125 Modena, Italy; (A.A.); (L.C.); (R.S.)
| | - Gigliola Lusvardi
- DSCG, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy;
- Correspondence: ; Tel.: +39-059-205-8549
| |
Collapse
|
90
|
Simfia I, Schiavi J, McNamara LM. Alterations in osteocyte mediated osteoclastogenesis during estrogen deficiency and under ROCK-II inhibition: An in vitro study using a novel postmenopausal multicellular niche model. Exp Cell Res 2020; 392:112005. [PMID: 32330507 DOI: 10.1016/j.yexcr.2020.112005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 01/03/2023]
Abstract
This study sought to derive an enhanced understanding of the complex intracellular interactions that drive bone loss in postmenopausal osteoporosis. We applied an in-vitro multicellular niche to recapitulate cell-cell signalling between osteocytes, osteoblasts and osteoclasts to investigate (1) how estrogen-deficient and mechanically loaded osteocytes regulate osteoclastogenesis and (2) whether ROCK-II inhibition affects these mechanobiological responses. We report that mechanically stimulated and estrogen-deficient osteocytes upregulated RANKL/OPG and M-CSF gene expression, when compared to those treated with 10 nM estradiol. Osteoclast precursors (RAW 264.7) cultured within this niche underwent significant reduction in osteoclastogenic gene expression (CTSK), and there was an increasing trend in the area covered by TRAP+ osteoclasts (24% vs. 19.4%, p = 0.06). Most interestingly, upon treatment with the ROCK-II inhibitor, RANKL/OPG and M-CSF gene expression by estrogen-deficient osteocytes were downregulated. Yet, this inhibition of the pro-osteoclastogenic factors by osteocytes did not ultimately reduce the differentiation of osteoclast precursors. Indeed, TRAP and CTSK gene expressions in osteoclast precursors were upregulated, and there was an increased trend for osteoclast area (30.4% vs. 24%, p = 0.07), which may have been influenced by static osteoblasts (MC3T3-E1) that were included in the niche. We conclude that ROCK-II inhibition can attenuate bone loss driven by osteocytes during estrogen deficiency.
Collapse
Affiliation(s)
- Irene Simfia
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Jessica Schiavi
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Laoise M McNamara
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
91
|
Parmentier L, Riffault M, Hoey DA. Utilizing Osteocyte Derived Factors to Enhance Cell Viability and Osteogenic Matrix Deposition within IPN Hydrogels. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E1690. [PMID: 32260406 PMCID: PMC7178658 DOI: 10.3390/ma13071690] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022]
Abstract
Many bone defects arising due to traumatic injury, disease, or surgery are unable to regenerate, requiring intervention. More than four million graft procedures are performed each year to treat these defects making bone the second most commonly transplanted tissue worldwide. However, these types of graft suffer from a limited supply, a second surgical site, donor site morbidity, and pain. Due to the unmet clinical need for new materials to promote skeletal repair, this study aimed to produce novel biomimetic materials to enhance stem/stromal cell osteogenesis and bone repair by recapitulating aspects of the biophysical and biochemical cues found within the bone microenvironment. Utilizing a collagen type I-alginate interpenetrating polymer network we fabricated a material which mirrors the mechanical and structural properties of unmineralized bone, consisting of a porous fibrous matrix with a young's modulus of 64 kPa, both of which have been shown to enhance mesenchymal stromal/stem cell (MSC) osteogenesis. Moreover, by combining this material with biochemical paracrine factors released by statically cultured and mechanically stimulated osteocytes, we further mirrored the biochemical environment of the bone niche, enhancing stromal/stem cell viability, differentiation, and matrix deposition. Therefore, this biomimetic material represents a novel approach to promote skeletal repair.
Collapse
Affiliation(s)
- Laurens Parmentier
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2 D02 DK07, Ireland
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Mathieu Riffault
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2 D02 DK07, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2 D02 VN51, Ireland
| | - David A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2 D02 DK07, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2 D02 VN51, Ireland
| |
Collapse
|
92
|
Storlino G, Colaianni G, Sanesi L, Lippo L, Brunetti G, Errede M, Colucci S, Passeri G, Grano M. Irisin Prevents Disuse-Induced Osteocyte Apoptosis. J Bone Miner Res 2020; 35:766-775. [PMID: 31826311 DOI: 10.1002/jbmr.3944] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
Abstract
Previous results showed that intermittently administered irisin improves bone mass in normal mice and prevents the development of disuse-induced osteoporosis and muscular atrophy in hindlimb-suspended mice, a murine model able to mimic the absence of mechanical loading. A recent study showed that irisin increases survival of osteocytes acting through integrin αV/β5 receptors. To better understand the action of irisin on these cells, we investigated the downstream signaling cascades in osteocyte-like cells (MLO-Y4) treated with recombinant irisin (rec-irisin) in vitro and we analyzed survival of osteocytes and caspase activation in cortical bone of osteoporotic mice treated with rec-irisin in vivo. Our results revealed that rec-irisin activated the MAP kinases Erk1 and Erk2 and increased the expression of the transcription factor Atf4 (2.5-fold, p < .05) through an Erk-dependent pathway in osteocytes. Some key genes expressed by MLO-Y4 cells were modulated by long-term irisin treatment, either continuously administered or given with intermittent short pulses. Interestingly, Sost mRNA was severely downregulated only upon intermittent irisin administration (10-fold, p < .001). Furthermore, rec-irisin upregulated Tfam mRNA (fourfold, p < .05) and Bcl2/Bax ratio (twofold, p < .05) in MLO-Y4 cells. By detecting caspase-9 and caspase-3, we also found that rec-irisin inhibited apoptosis induced by hydrogen peroxide and dexamethasone, respectively. In cortical bone of unloading C57BL6 mice treated with vehicle (unload-veh), irisin prevented disuse-induced reduction of viable osteocytes (+30% versus unload-veh, p < .05) and increase of empty lacunae (+110% versus unload-veh, p < .05), as well as caspase-9 (threefold, p < .05) and caspase-3 (twofold, p < .05) activations. Our findings revealed underlying mechanisms of irisin action on osteocytes, which increases their functions and exerts anti-apoptotic effects, confirming that mechanosensor cells of bone are sensitive to the exercise-mimetic myokine irisin. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Giuseppina Storlino
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Graziana Colaianni
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Luciana Lippo
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Silvia Colucci
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Giovanni Passeri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
93
|
Moor MB, Bonny O. Memo1 gene expression in kidney and bone is unaffected by dietary mineral load and calciotropic hormones. Physiol Rep 2020; 8:e14410. [PMID: 32291966 PMCID: PMC7156332 DOI: 10.14814/phy2.14410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
Mediator of cell motility 1 (MEMO1) is a ubiquitously expressed modulator of cellular responses to growth factors including FGF23 signaling, and Memo1-deficient mice share some phenotypic traits with Fgf23- or Klotho-deficient mouse models. Here, we tested whether Memo1 gene expression is regulated by calciotropic hormones or by changing the dietary mineral load. MLO-Y4 osteocyte-like cells were cultured and treated with 1,25(OH)2 -vitamin D3 . Wild-type C57BL/6N mice underwent treatments with 1,25(OH)2 -vitamin D3 , parathyroid hormone, 17β-estradiol or vehicle. Other cohorts of C57BL/6N mice were fed diets varying in calcium or phosphate content. Expression of Memo1 and control genes was assessed by qPCR. 1,25(OH)2 -vitamin D3 caused an acute decrease in Memo1 transcript levels in vitro, but not in vivo. None of the hormones tested had an influence on Memo1 transcripts, whereas the assessed control genes reacted the expected way. Dietary interventions with calcium and phosphate did not affect Memo1 transcripts but altered the chosen control genes' expression. We observed that Memo1 was not regulated by calciotropic hormones or change in mineral load, suggesting major differences between the regulation and physiological roles of Klotho, Fgf23, and Memo1.
Collapse
Affiliation(s)
- Matthias B. Moor
- Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
- The National Centre of Competence in Research (NCCR) "Kidney.CH ‐ Kidney Control of Homeostasis" SwitzerlandZürichSwitzerland
- Present address:
Department of Nephrology and HypertensionBern University HospitalBernSwitzerland
| | - Olivier Bonny
- Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
- The National Centre of Competence in Research (NCCR) "Kidney.CH ‐ Kidney Control of Homeostasis" SwitzerlandZürichSwitzerland
- Service of NephrologyDepartment of MedicineLausanne University HospitalLausanneSwitzerland
| |
Collapse
|
94
|
Iyer S, Melendez-Suchi C, Han L, Baldini G, Almeida M, Jilka RL. Elevation of the unfolded protein response increases RANKL expression. FASEB Bioadv 2020; 2:207-218. [PMID: 32259048 PMCID: PMC7133738 DOI: 10.1096/fba.2019-00032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/12/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increased production of the osteoclastogenic cytokine RANKL is a common feature of pathologic bone loss, but the underlying cause of this increase is poorly understood. The unfolded protein response (UPR) is activated in response to accumulation of misfolded proteins in the endoplasmic reticulum (ER). Failure to resolve misfolding results in excess UPR signaling that stimulates cytokine production and cell death. We therefore investigated whether RANKL is one of the cytokines stimulated in response to elevated UPR in bone cells. Pharmacologic induction of UPR with tunicamycin (Tm)-stimulated RANKL expression in cultures of primary osteoblastic cells and in osteoblast and osteocyte cell lines. Pharmacologic inhibition of the UPR blunted Tm-induced RANKL production. Silencing Edem1 or Sel1l, proteins that aid in degradation of misfolded proteins, also induced UPR and increased RANKL mRNA. Moreover, Tm or hypoxia increased RANKL and bone resorption in cultures of neonatal murine calvaria. Administration of Tm to adult mice caused dilation of ER in osteoblasts and osteocytes, elevated the UPR, and increased RANKL expression and osteoclast number. These findings support the hypothesis that excessive UPR signaling stimulates the expression of RANKL by osteoblasts and osteocytes, and thereby facilitates excessive bone resorption and bone loss in pathologic conditions.
Collapse
Affiliation(s)
- Srividhya Iyer
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
| | | | - Li Han
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology University of Arkansas Medical Sciences Little Rock AR USA
| | - Maria Almeida
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
- Central Arkansas Veterans Healthcare System Little Rock AR USA
| |
Collapse
|
95
|
Saunders RK, Infanti J, Ali H, Shuey T, Potteiger C, McNeilly S, Adams CS. Gram-Negative Bacteria Are Internalized Into Osteocyte-Like Cells. J Orthop Res 2020; 38:861-870. [PMID: 31692074 DOI: 10.1002/jor.24510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/14/2019] [Indexed: 02/04/2023]
Abstract
While Gram-positive organisms are the most common causative agent of initial bone infections, the percentage of Gram-negative species increases in reoccurring bone infections. As bacterial internalization has been suggested as one cause of reoccurring bone infection, we tested the hypothesis that Gram-negative species of bacteria can be internalized into bone cells. Using the MLO-A5 and the MLO-Y4 cell lines as our cell models, we demonstrated that the Gram-negative species, Proteus mirabilis and Serratia marcescens, can be internalized in these cells using an internalization assay. This rate at which these two species were internalized was both time- and initial concentration-dependent. Confocal analysis demonstrated the presence of internalized bacteria within both cell types. Inhibition of the cellular uptake with methyl-β-cyclodextrin and chloroquine both reduced internalized bacteria, indicating that this process is, at least in part, cell mediated. Finally, we demonstrated that the presence of internalized P. mirabilis did not impact cell viability, measured either by lactate dehydrogenase (LDH) release or 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) activity, while the presence of S. marcescens, on the other hand, both increased LDH release and reduced MTT activity, indicating a loss of cell viability in response to the organism. These results indicated that both species of Gram-negative bacteria can be internalized by bone cells and that these internalized bacteria could potentially result in reoccurring bone infections. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:861-870, 2020.
Collapse
Affiliation(s)
- Ray K Saunders
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph Infanti
- Graduate School of Biological Sciences, Rowan University, Stratford, New Jersey
| | - Hibah Ali
- Touro College of Osteopathic Medicine, New York, New York
| | - Timothy Shuey
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Courtney Potteiger
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Shelby McNeilly
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Christopher S Adams
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
96
|
Aziz AH, Wilmoth RL, Ferguson VL, Bryant SJ. IDG-SW3 Osteocyte Differentiation and Bone Extracellular Matrix Deposition Are Enhanced in a 3D Matrix Metalloproteinase-Sensitive Hydrogel. ACS APPLIED BIO MATERIALS 2020; 3:1666-1680. [PMID: 32719827 DOI: 10.1021/acsabm.9b01227] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteocytes reside within a heavily mineralized matrix making them difficult to study in vivo and to extract for studies in vitro. IDG-SW3 cells are capable of producing mineralized collagen matrix and transitioning from osteoblasts to mature osteocytes, thus offering an alternative to study osteoblast to late osteocyte differentiation in vitro. The goal for this work was to develop a 3D degradable hydrogel to support IDG-SW3 differentiation and deposition of bone ECM. In 2D, the genes Mmp2 and Mmp13 increased during IDG-SW3 differentiation and were used as targets to create a MMP-sensitive poly(ethylene glycol) hydrogel containing the peptide crosslink GCGPLG-LWARCG and RGD to promote cell attachment. IDG-SW3 differentiation in the MMP-sensitive hydrogels improved over non-degradable hydrogels and standard 2D culture. Alkaline phosphatase activity at day 14 was higher, Dmp1 and Phex were 8.1-fold and 3.8-fold higher, respectively, and DMP1 protein expression was more pronounced in the MMP-sensitive hydrogels compared to non-degradable hydrogels. Cell-encapsulation density (cells/ml precursor) influenced formation of dendrite-like cellular process and mineral and collagen deposition with 80×106 performing better than 2×106 or 20×106, while connexin 43 was not affected by cell density. The cell density effects were more pronounced in the MMP-sensitive hydrogels over non-degradable hydrogels. This study identified that high cell encapsulation density and a hydrogel susceptible to cell-mediated degradation enhanced mineralized collagen matrix and osteocyte differentiation. Overall, a promising hydrogel is presented that supports IDG-SW3 cell maturation from osteoblasts to osteocytes in 3D.
Collapse
Affiliation(s)
- Aaron H Aziz
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA.,BioFrontiers Institute, University of Colorado, Boulder, CO 80309 USA
| | - Rachel L Wilmoth
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309 USA
| | - Virginia L Ferguson
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309 USA.,Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309 USA.,Material Science and Engineering, University of Colorado, Boulder, CO 80309 USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO USA.,BioFrontiers Institute, University of Colorado, Boulder, CO 80309 USA.,Material Science and Engineering, University of Colorado, Boulder, CO 80309 USA
| |
Collapse
|
97
|
Bellido T, Delgado-Calle J. Ex Vivo Organ Cultures as Models to Study Bone Biology. JBMR Plus 2020; 4:JBM410345. [PMID: 32161838 PMCID: PMC7059827 DOI: 10.1002/jbm4.10345] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
The integrity of the skeleton is maintained by the coordinated and balanced activities of the bone cells. Osteoclasts resorb bone, osteoblasts form bone, and osteocytes orchestrate the activities of osteoclasts and osteoblasts. A variety of in vitro approaches has been used in an attempt to reproduce the complex in vivo interactions among bone cells under physiological as well as pathological conditions and to test new therapies. Most cell culture systems lack the proper extracellular matrix, cellular diversity, and native spatial distribution of the components of the bone microenvironment. In contrast, ex vivo cultures of fragments of intact bone preserve key cell-cell and cell-matrix interactions and allow the study of bone cells in their natural 3D environment. Further, bone organ cultures predict the in vivo responses to genetic and pharmacologic interventions saving precious time and resources. Moreover, organ cultures using human bone reproduce human conditions and are a useful tool to test patient responses to therapeutic agents. Thus, these ex vivo approaches provide a platform to perform research in bone physiology and pathophysiology. In this review, we describe protocols optimized in our laboratories to establish ex vivo bone organ cultures and provide technical hints and suggestions. In addition, we present examples on how this technical approach can be employed to study osteocyte biology, drug responses in bone, cancer-induced bone disease, and cross-talk between bone and other organs © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Teresita Bellido
- Department of Anatomy, Cell Biology & Physiology Indiana University School of Medicine Indianapolis IN USA.,Division of Endocrinology, Department of Medicine Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Richard L. Roudebush Veterans Affairs Medical Center Indianapolis IN USA
| | - Jesus Delgado-Calle
- Department of Anatomy, Cell Biology & Physiology Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Richard L. Roudebush Veterans Affairs Medical Center Indianapolis IN USA.,Division of Hematology/Oncology, Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| |
Collapse
|
98
|
Creecy A, Damrath JG, Wallace JM. Control of Bone Matrix Properties by Osteocytes. Front Endocrinol (Lausanne) 2020; 11:578477. [PMID: 33537002 PMCID: PMC7848033 DOI: 10.3389/fendo.2020.578477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Osteocytes make up 90-95% of the cellular content of bone and form a rich dendritic network with a vastly greater surface area than either osteoblasts or osteoclasts. Osteocytes are well positioned to play a role in bone homeostasis by interacting directly with the matrix; however, the ability for these cells to modify bone matrix remains incompletely understood. With techniques for examining the nano- and microstructure of bone matrix components including hydroxyapatite and type I collagen becoming more widespread, there is great potential to uncover novel roles for the osteocyte in maintaining bone quality. In this review, we begin with an overview of osteocyte biology and the lacunar-canalicular system. Next, we describe recent findings from in vitro models of osteocytes, focusing on the transitions in cellular phenotype as they mature. Finally, we describe historical and current research on matrix alteration by osteocytes in vivo, focusing on the exciting potential for osteocytes to directly form, degrade, and modify the mineral and collagen in their surrounding matrix.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
| | - John G. Damrath
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
- *Correspondence: Joseph M. Wallace,
| |
Collapse
|
99
|
Ueda S, Shimasaki M, Ichiseki T, Hirata H, Kawahara N, Ueda Y. Mitochondrial Transcription Factor A added to Osteocytes in a Stressed Environment has a Cytoprotective Effect. Int J Med Sci 2020; 17:1293-1299. [PMID: 32547324 PMCID: PMC7294917 DOI: 10.7150/ijms.45335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/06/2020] [Indexed: 02/03/2023] Open
Abstract
The main precipitant of glucocorticoid-associated femoral head osteonecrosis is widely accepted to be an ischemic-hypoxic event, with oxidative stress also as an underlying factor. Mitochondrial DNA is more vulnerable to oxidative injury than the nucleus, and mitochondrial transcription factor A (TFAM), which plays roles in its function, preservation, and regulation is being increasingly investigated. In the present study we focused on the impact of TFAM on the relation between the oxidative injury induced by the addition of glucocorticoid to a hypoxic environment and osteocytic cell necrosis. Using cultured osteocytes MLO-Y4 in a 1% hypoxic environment (hypoxia) to which 1µM dexamethasone (Dex) was added (Dex(+)/hypoxia(+)), an immunocytochemical study was conducted using 8-hydroxy-2'-deoxyguanosine (8-OHdG), an index of oxidative stress, and hypoxia inducible factor-1α (HIF-1α), a marker of hypoxia. Next, after adding TFAM siRNA, TFAM knockdown, cultured for 24h, and mitochondrial membrane potential were measured, they were stained with ATP5A which labels adenosine triphosphate (ATP) production. Dex was added to MLO-Y4 to which TFAM had been added, and cultured for 24h in hypoxia. The ratio of dead cells to viable cells was determined and compared. Enhanced expression of 8-OHdG, HIF-1α was found in osteocytes following the addition of glucocorticoid in a hypoxic environment. With TFAM knockdown, as compared to normoxia, mitochondrial function significantly decreased. On the other hand, by adding TFAM, the incidence of osteocytic cell necrosis was significantly decreased as compared with Dex(+)/hypoxia(+). TFAM was confirmed to be important in mitochondrial function and preservation, inhibition of oxidative injury and maintenance of ATP production. Moreover, prevention of mitochondrial injury can best be achieved by decreasing the development of osteocytic cell necrosis.
Collapse
Affiliation(s)
- Shusuke Ueda
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Miyako Shimasaki
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| | - Toru Ichiseki
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Hiroaki Hirata
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Norio Kawahara
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Yoshimichi Ueda
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| |
Collapse
|
100
|
Xie J, Guo J, Kanwal Z, Wu M, Lv X, Ibrahim NA, Li P, Buabeid MA, Arafa ESA, Sun Q. Calcitonin and Bone Physiology: In Vitro, In Vivo, and Clinical Investigations. Int J Endocrinol 2020; 2020:3236828. [PMID: 32963524 PMCID: PMC7501564 DOI: 10.1155/2020/3236828] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Calcitonin was discovered as a peptide hormone that was known to reduce the calcium levels in the systemic circulation. This hypocalcemic effect is produced due to multiple reasons such as inhibition of bone resorption or suppression of calcium release from the bone. Thus, calcitonin was said as a primary regulator of the bone resorption process. This is the reason why calcitonin has been used widely in clinics for the treatment of bone disorders such as osteoporosis, hypercalcemia, and Paget's disease. However, presently calcitonin usage is declined due to the development of efficacious formulations of new drugs. Calcitonin gene-related peptides and several other peptides such as intermedin, amylin, and adrenomedullin (ADM) are categorized in calcitonin family. These peptides are known for the structural similarity with calcitonin. Aside from having a similar structure, these peptides have few overlapping biological activities and signal transduction action through related receptors. However, several other activities are also present that are peptide specific. In vitro and in vivo studies documented the posttreatment effects of calcitonin peptides, i.e., positive effect on bone osteoblasts and their formation and negative effect on osteoclasts and their resorption. The recent research studies carried out on genetically modified mice showed the inhibition of osteoclast activity by amylin, while astonishingly calcitonin plays its role by suppressing osteoblast and bone turnover. This article describes the review of the bone, the activity of the calcitonin family of peptides, and the link between them.
Collapse
Affiliation(s)
- Jingbo Xie
- Department of Orthopedics, Fengcheng People's Hospital, Fengcheng, Jiangxi 331100, China
| | - Jian Guo
- Department of the Second Orthopedics, Hongdu Hospital of Traditional Chinese Medicine Affiliated to Jiangxi University of Traditional Chinese Medicine, Nanchang Hongdu Traditional Chinese Medicine Hospital, Nanchang, Jiangxi 330008, China
| | | | - Mingzheng Wu
- Department of Orthopaedics, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xiangyang Lv
- Department of Orthopaedics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China
| | | | - Ping Li
- Department of Orthopaedics, Ya'an People's Hospital, Ya'an, Sichuan 625000, China
| | | | | | - Qingshan Sun
- Department of Orthopedics, The Third Hospital of Shandong Province, Jinan, Shandong 250031, China
| |
Collapse
|