1
|
Hoveidaei AH, Sadat-Shojai M, Nabavizadeh SS, Niakan R, Shirinezhad A, MosalamiAghili S, Tabaie S. Clinical challenges in bone tissue engineering - A narrative review. Bone 2025; 192:117363. [PMID: 39638083 DOI: 10.1016/j.bone.2024.117363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Bone tissue engineering (BTE) has emerged as a promising approach to address large bone defects caused by trauma, infections, congenital malformations, and tumors. This review focuses on scaffold design, cell sources, growth factors, and vascularization strategies, highlighting their roles in developing effective treatments. We explore the complexities of balancing mechanical properties, porosity, and biocompatibility in scaffold materials, alongside optimizing mesenchymal stem cell delivery methods. The critical role of growth factors in bone regeneration and the need for controlled release systems are discussed. Vascularization remains a significant hurdle, with strategies such as angiogenic factors, co-culture systems, and bioprinting under investigation. Mechanical challenges, tissue responses, and inflammation management are examined, alongside gene therapy's potential for enhancing osteogenesis and angiogenesis via both viral and non-viral delivery methods. The review emphasizes the impact of patient-specific factors on bone healing outcomes and the importance of personalized approaches. Future directions are described, emphasizing the necessity of interdisciplinary cooperation to advance the field of BTE and convert laboratory results into clinically feasible solutions.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Sara S Nabavizadeh
- Otolaryngology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Niakan
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Sean Tabaie
- Department of Orthopaedic Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
2
|
Huang G, Yin W, Zhao X, Xu M, Wang P, Li R, Zhou L, Tang W, Jiao J. Osteoking inhibits apoptosis of BMSCs in osteoporotic rats via PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:118961. [PMID: 39653105 DOI: 10.1016/j.jep.2024.118961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 01/04/2025]
Abstract
In China, Osteoking is a commonly used treatment and preventive measure for osteoporosis. The pathophysiology of osteoporosis is closely associated with apoptosis; however, it remains unclear whether the role of Osteoking in promoting bone formation is linked to apoptosis. AIM OF STUDY This study aims to investigate whether Osteoking inhibits apoptosis of BMSCs in osteoporotic rats via the PI3K/AKT signaling pathway and to conduct a detailed exploration of this mechanism. The goal is to provide a theoretical basis for the clinical application of Osteoking in osteoporosis treatment. METHODS A rat model of osteoporosis was established through bilateral ovariectomy (OVX), followed by treatment with Osteoking. After ten weeks of therapy, BMD was evaluated. The biomechanics of the left tibia were measured, the left femur was sequenced, and the right tibia was stained using histomorphometric and Masson's staining methods. Peripheral serum was collected to measure bone-related markers, including E2, PINP, and CTX. RNA-Seq results were verified using the remaining bone samples. Comparative analysis demonstrated the efficacy of Osteoking in treating osteoporosis and provided preliminary insights into the underlying mechanisms. Primary BMSCs were cultured using bone marrow apposition. CCK8 assays were conducted to screen the intervention conditions of Osteoking and LY294002. Various concentrations of Osteoking-containing serum and LY294002 were tested separately to determine the optimal intervention concentration for drug delivery. The impact of Osteoking on lipid formation was also evaluated. Following treatment of BMSCs from OVX rats with Sham serum, OVX serum, OVX + LY294002 serum, and Osteoking + LY294002 serum, the expression of PI3K/AKT/mTOR, osteogenesis-related regulatory factors, and apoptosis-related regulatory factors was assessed. Flow cytometry was employed to evaluate apoptosis in BMSCs. RESULTS Osteoking significantly improved whole-body BMD and bone biomechanical indices in OVX rats. It also significantly elevated the serum levels of E2 and PINP while reducing the level of CTX, which significantly improved bone microstructure and promoted new bone formation. RNA-seq analysis indicated that the therapeutic mechanism involved the PI3K/AKT signaling pathway. Osteoking increased the expression of RUNX2 and decreased the expression of PPAR-γ, a marker of lipogenesis, in OVX rats. Extraction of BMSCs for subsequent studies revealed a significant reduction in proliferation and osteogenic differentiation, along with an increase in lipogenic differentiation, in the OVX group. Osteoking treatment inhibited the expression of PPAR-γ and increased the expression of RUNX2 in BMSCs. Additionally, Osteoking reversed the LY294002-mediated inhibition of PI3K/AKT/mTOR signaling pathway activation, increased the expression of the apoptosis-protecting protein Bcl2, and decreased the expression of apoptosis-associated proteins Caspase3 and Bax. CONCLUSION Osteoking markedly improved bone microstructure, biomechanics, and bone density in OVX rats. Osteoking-containing serum reversed the imbalance in lineage differentiation in OVX rats, characterized by reduced osteogenic differentiation and increased lipid differentiation of BMSCs. Furthermore, Osteoking-containing serum significantly increased BMSC proliferation and prevented apoptosis in OVX rats through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Guijiang Huang
- Department of Science and Education, The First Affiliated Hospital of Kunming Medical University, Kunming, 650600, China; Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650600, China
| | - Wenjie Yin
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650600, China
| | - Xin Zhao
- Department of Science and Education, The First Affiliated Hospital of Kunming Medical University, Kunming, 650600, China
| | - Muli Xu
- Kunming Medical University, Kunming, 650600, China
| | - Peijin Wang
- Kunming Medical University, Kunming, 650600, China
| | - Rong Li
- Kunming Medical University, Kunming, 650600, China
| | - Li Zhou
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650600, China
| | - Wei Tang
- Department of Science and Education, The First Affiliated Hospital of Kunming Medical University, Kunming, 650600, China.
| | - Jianlin Jiao
- Kunming Medical University, Kunming, 650600, China.
| |
Collapse
|
3
|
Zhou W, Zhu C, Zhou F. TXNIP mediated by EZH2 regulated osteogenic differentiation in hBmscs and MC3T3-E1 cells through the modulation of oxidative stress and PI3K/AKT/Nrf2 pathway. Connect Tissue Res 2024; 65:293-303. [PMID: 38884152 DOI: 10.1080/03008207.2024.2358361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Previous research has identified a significant role of Thioredoxin-interacting protein (TXNIP) in bone loss. The purpose of this investigation was to assess the role and the underlying molecular mechanisms of TXNIP in the osteogenic differentiation of human bone marrow stromal cells (hBMSCs) and pre-osteoblast MC3T3-E1 cells. METHODS Human bone marrow stem cells (hBMSCs) and MC3T3-E1 cells were used to induce osteogenic differentiation. The expression of genes and proteins was assessed using RT-qPCR and western blot, respectively. ChIP assay was used to validate the interaction between genes. The osteogenic differentiation ability of cells was reflected using ALP staining and detection of ALP activity. The mineralization ability of cells was assessed using ARS staining. DCFCA staining was employed to evaluate the intracellular ROS level. RESULTS Initially, downregulation of TXNIP and upregulation of EZH2 were observed during osteogenesis in hBMSCs and MC3T3-E1 cells. Additionally, it was discovered that EZH2 negatively regulates TXNIP expression in these cells. Furthermore, experiments indicated that the knockdown of TXNIP stimulated the activation of the PI3K/AKT/Nrf2 signaling pathway in hBMSCs and MC3T3- E1 cells, thus inhibiting the production of reactive oxygen species (ROS). Further functional experiments revealed that overexpression of TXNIP inhibited the osteogenic differentiation in hBMSCs and MC3T3-E1 cells by enhancing ROS produc-tion. On the other hand, knockdown of TXNIP promoted the osteogenic differentiation capacity of hBMSCs and MC3T3-E1 cells through the activation of the PI3K/AKT/Nrf2 pathway. CONCLUSION In conclusion, this study demonstrated that TXNIP expression, under the regulation of EZH2, plays a crucial role in the osteogenic differentiation of hBMSCs and MC3T3-E1 cells by regulating ROS production and the PI3K/AKT/Nrf2 pathway.
Collapse
Affiliation(s)
- Weibo Zhou
- Department of Orthopedics, Changzhou No. 2 People's Hospital, Changzhou, China
| | - Chunhui Zhu
- Department of Orthopedics, Changzhou No. 2 People's Hospital, Changzhou, China
| | - Fulin Zhou
- Department of Orthopedics, Changzhou No. 2 People's Hospital, Changzhou, China
| |
Collapse
|
4
|
Song J, Wang Y, Zhu Z, Wang W, Yang H, Shan Z. Negative Regulation of LINC01013 by METTL3 and YTHDF2 Enhances the Osteogenic Differentiation of Senescent Pre-Osteoblast Cells Induced by Hydrogen Peroxide. Adv Biol (Weinh) 2024; 8:e2300642. [PMID: 38548669 DOI: 10.1002/adbi.202300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/19/2024] [Indexed: 05/15/2024]
Abstract
Senescent pre-osteoblasts have a reduced ability to differentiate, which leads to a reduction in bone formation. It is critical to identify the keys that regulate the differentiation fate of senescent pre-osteoblasts. LINC01013 has an essential role in cell stemness, differentiation, and senescence regulation. This study aims to examine the role and mechanism of LINC01013 in regulating osteogenic differentiation in senescent human embryonic osteoblast cell line (hFOB1.19) cells induced by hydrogen peroxide (H2O2). The results show that LINC01013 decreased alkaline phosphatase activity, mineralization of hFOB1.19 cells in vitro, and the expression of collagen II, osteocalcin, and bone sialoprotein. LINC01013 knockdown enhances the osteogenesis of hFOB1.19 cells and rescues osteogenic differentiation impaired by H2O2. METTL3 negatively regulates LINC01013 expression, enhancing hFOB1.19 cells' osteogenesis in vitro and in vivo. METTL3 overexpression can enhance hFOB1.19 cells' osteogenic differentiation impaired by H2O2. YTHDF2 promotes LINC01013 decay, facilitating osteogenic differentiation. YTHDF2 overexpression rescues hFOB1.19 cells osteogenic differentiation impaired by H2O2. Taken together, METTL3 upregulates osteogenic differentiation by inhibiting LINC01013, and YTHDF2 accelerates LINC01013 degradation, reducing its inhibitory effect. This study highlights LINC01013 as a key regulator in the fate switching process of senescent hFOB1.19 cells, impacting osteogenic differentiation.
Collapse
Affiliation(s)
- Jiaxin Song
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Yuejun Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Zhao Zhu
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Wanqing Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Zhaochen Shan
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, 100050, China
| |
Collapse
|
5
|
Yılmaz D, Mathavan N, Wehrle E, Kuhn GA, Müller R. Mouse models of accelerated aging in musculoskeletal research for assessing frailty, sarcopenia, and osteoporosis - A review. Ageing Res Rev 2024; 93:102118. [PMID: 37935249 DOI: 10.1016/j.arr.2023.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Musculoskeletal aging encompasses the decline in bone and muscle function, leading to conditions such as frailty, osteoporosis, and sarcopenia. Unraveling the underlying molecular mechanisms and developing effective treatments are crucial for improving the quality of life for those affected. In this context, accelerated aging models offer valuable insights into these conditions by displaying the hallmarks of human aging. Herein, this review focuses on relevant mouse models of musculoskeletal aging with particular emphasis on frailty, osteoporosis, and sarcopenia. Among the discussed models, PolgA mice in particular exhibit hallmarks of musculoskeletal aging, presenting early-onset frailty, as well as reduced bone and muscle mass that closely resemble human musculoskeletal aging. Ultimately, findings from these models hold promise for advancing interventions targeted at age-related musculoskeletal disorders, effectively addressing the challenges posed by musculoskeletal aging and associated conditions in humans.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland; AO Research Institute Davos, Davos Platz, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Yusop N, Moseley R, Waddington RJ. Hyperglycemia exerts disruptive effects on the secretion of TGF-β 1 and its matrix ligands, decorin and biglycan, by mesenchymal sub-populations and macrophages during bone repair. FRONTIERS IN DENTAL MEDICINE 2023; 4:1200122. [PMID: 39916897 PMCID: PMC11797960 DOI: 10.3389/fdmed.2023.1200122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/05/2023] [Indexed: 02/09/2025] Open
Abstract
Introduction Bone has a high capacity for repair, but for patients with uncontrolled type 2 diabetes mellitus (T2DM), the associated hyperglycemia can significantly delay osteogenic processes. These patients respond poorly to fracture repair and bone grafts, leading to lengthy care plans due to arising complications. Mesenchymal stromal cells (MSCs) and M2 macrophages are both major sources of transforming growth factor-β1 (TGF-β1), a recognized mediator for osteogenesis and whose bioavailability and activities are further regulated by matrix small leucine-rich proteoglycans (SLRPs), decorin and biglycan. The aim of this study was to investigate how in vivo and in vitro hyperglycemic (HGly) environments can influence the levels of TGF-β1, decorin, and biglycan during bone repair, with additional consideration for how long-term glucose exposure and cell aging can also influence this process. Results Following bone healing within a T2DM in vivo model, histological and immunolabeling analyses of bone tissue sections confirmed delayed healing, which was associated with significantly elevated TGF-β1 levels within the bone matrices of young diabetic rats, compared with their normoglycemic (Norm) and aged counterparts. Studies continued to assess in vitro the effects of normal (5.5 mM) and high (25 mM) glucose exposure on the osteogenic differentiation of compact bone-derived mesenchymal stromal cells (CB-MSCs) at population doubling (PD)15, characterized to contain populations of lineage-committed osteoblasts, and at PD150, where transit-amplifying cells predominate. Short-term glucose exposure increased TGF-β1 and decorin secretion by committed osteoblasts but had a lesser effect on transit-amplifying cells. In contrast, the long-term exposure of CB-MSCs to high glucose was associated with decreased TGF-β1 and increased decorin secretion. Similar assessments on macrophage populations indicated high glucose inhibited TGF-β1 secretion, preventing M2 formation. Discussion Collectively, these findings highlight how hyperglycemia associated with T2DM can perturb TGF-β1 and decorin secretion by MSCs and macrophages, thereby potentially influencing TGF-β1 bioavailability and signaling during bone repair.
Collapse
Affiliation(s)
- Norhayati Yusop
- School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | | | | |
Collapse
|
7
|
Elmansi AM, Eisa NH, Periyasamy-Thandavan S, Kondrikova G, Kondrikov D, Calkins MM, Aguilar-Pérez A, Chen J, Johnson M, Shi XM, Reitman C, McGee-Lawrence ME, Crawford KS, Dwinell MB, Volkman BF, Blumer JB, Luttrell LM, McCorvy JD, Hill WD. DPP4-Truncated CXCL12 Alters CXCR4/ACKR3 Signaling, Osteogenic Cell Differentiation, Migration, and Senescence. ACS Pharmacol Transl Sci 2023; 6:22-39. [PMID: 36659961 PMCID: PMC9844133 DOI: 10.1021/acsptsci.2c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Bone marrow skeletal stem cells (SSCs) secrete many cytokines including stromal derived factor-1 or CXCL12, which influences cell proliferation, migration, and differentiation. All CXCL12 splice variants are rapidly truncated on their N-terminus by dipeptidyl peptidase 4 (DPP4). This includes the common variant CXCL12 alpha (1-68) releasing a much less studied metabolite CXCL12(3-68). Here, we found that CXCL12(3-68) significantly inhibited SSC osteogenic differentiation and RAW-264.7 cell osteoclastogenic differentiation and induced a senescent phenotype in SSCs. Importantly, pre-incubation of SSCs with CXCL12(3-68) significantly diminished their ability to migrate toward CXCL12(1-68) in transwell migration assays. Using a high-throughput G-protein-coupled receptor (GPCR) screen (GPCRome) and bioluminescent resonance energy transfer molecular interaction assays, we revealed that CXCL12(3-68) acts via the atypical cytokine receptor 3-mediated β-arrestin recruitment and as a competitive antagonist to CXCR4-mediated signaling. Finally, a reverse phase protein array assay revealed that DPP4-cleaved CXCL12 possesses a different downstream signaling profile from that of intact CXCL12 or controls. The data presented herein provides insights into regulation of CXCL12 signaling. Importantly, it demonstrates that DPP4 proteolysis of CXCL12 generates a metabolite with significantly different and previously overlooked bioactivity that helps explain discrepancies in the literature. This also contributes to an understanding of the molecular mechanisms of osteoporosis and bone fracture repair and could potentially significantly affect the interpretation of experimental outcomes with clinical consequences in other fields where CXCL12 is vital, including cancer biology, immunology, cardiovascular biology, neurobiology, and associated pathologies.
Collapse
Affiliation(s)
- Ahmed M. Elmansi
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Department of Pathology, University of
Michigan School of Medicine, Ann Arbor, Michigan 48109, United
States
| | - Nada H. Eisa
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Department of Biochemistry, Faculty of Pharmacy,
Mansoura University, Mansoura 35516,
Egypt
| | | | - Galina Kondrikova
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
| | - Maggie M. Calkins
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 W. Watertown Plank Road,
Milwaukee, Wisconsin 53226, United States
| | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology,
Indiana University School of Medicine in Indianapolis,
Indianapolis, Indiana 46202, United States
- Department of Cellular and Molecular Biology, School
of Medicine, Universidad Central Del Caribe, Bayamon, Puerto
Rico 00956, United States
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
| | - Jie Chen
- Division of Biostatistics and Data Science,
Department of Population Health Science, Medical College of Georgia, Augusta
University, Augusta, Georgia 30912, United States
| | - Maribeth Johnson
- Division of Biostatistics and Data Science,
Department of Population Health Science, Medical College of Georgia, Augusta
University, Augusta, Georgia 30912, United States
| | - Xing-ming Shi
- Department of Orthopaedic Surgery, Medical
College of Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Department of Neuroscience and Regenerative
Medicine, Medical College of Georgia, Augusta University,
Augusta, Georgia 30912, United States
| | - Charles Reitman
- Orthopaedics and Physical Medicine Department,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
| | - Meghan E. McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Department of Orthopaedic Surgery, Medical
College of Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Center for Healthy Aging, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
| | - Kyler S. Crawford
- Department of Biochemistry,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Michael B. Dwinell
- Department of Microbiology and Immunology,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Brian F. Volkman
- Department of Biochemistry,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Joe B. Blumer
- Department of Cell and Molecular Pharmacology and
Experimental Therapeutics, Medical University of South
Carolina, Charleston, South Carolina 29425, United
States
| | - Louis M. Luttrell
- Division of Endocrinology, Diabetes and
Medical Genetics, Medical University of South Carolina,
Charleston, South Carolina 29403, United States
| | - John D. McCorvy
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 W. Watertown Plank Road,
Milwaukee, Wisconsin 53226, United States
| | - William D. Hill
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Center for Healthy Aging, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Charlie Norwood Veterans Affairs
Medical Center, Augusta, Georgia 30904, United
States
| |
Collapse
|
8
|
Durham EL, Grey ZJ, Black L, Howie RN, Barth JL, Lee BS, Cray JJ. Sfrp4 expression in thyroxine treated calvarial cells. Life Sci 2022; 311:121158. [PMID: 36370870 PMCID: PMC9719041 DOI: 10.1016/j.lfs.2022.121158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
AIMS Evidence suggests alterations of thyroid hormone levels can disrupt normal bone development. Most data suggest the major targets of thyroid hormones to be the Htra1/Igf1 pathway. Recent discovery by our group suggests involvement of targets WNT pathway, specifically overexpression of antagonist Sfrp4 in the presence of exogenous thyroid hormone. MAIN METHODS Here we aimed to model these interactions in vitro using primary and isotype cell lines to determine if thyroid hormone drives increased Sfrp4 expression in cells relevant to craniofacial development. Transcriptional profiling, bioinformatics interrogation, protein and function analyses were used. KEY FINDINGS Affymetrix transcriptional profiling found Sfrp4 overexpression in primary cranial suture derived cells stimulated with thyroxine in vitro. Interrogation of the SFRP4 promoter identified multiple putative binding sites for thyroid hormone receptors. Experimentation with several cell lines demonstrated that thyroxine treatment induced Sfrp4 expression, demonstrating that Sfrp4 mRNA and protein levels are not tightly coupled. Transcriptional and protein analyses demonstrate thyroid hormone receptor binding to the proximal promoter of the target gene Sfrp4 in murine calvarial pre-osteoblasts. Functional analysis after thyroxine hormone stimulation for alkaline phosphatase activity shows that pre-osteoblasts increase alkaline phosphatase activity compared to other cell types, suggesting cell type susceptibility. Finally, we added recombinant SFRP4 to pre-osteoblasts in combination with thyroxine treatment and observed a significant decrease in alkaline phosphatase positivity. SIGNIFICANCE Taken together, these results suggest SFRP4 may be a key regulatory molecule that prevents thyroxine driven osteogenesis. These data corroborate clinical findings indicating a potential for SFRP4 as a diagnostic or therapeutic target for hyperostotic craniofacial disorders.
Collapse
Affiliation(s)
- Emily L Durham
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zachary J Grey
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Laurel Black
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - R Nicole Howie
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Jeremy L Barth
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Beth S Lee
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - James J Cray
- Department of Biomedical Education and Anatomy, College of Medicine, The Ohio State University, Columbus, OH, USA; Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
9
|
Yang C, Li Z, Liu Y, Hou R, Lin M, Fu L, Wu D, Liu Q, Li K, Liu C. Single-cell spatiotemporal analysis reveals cell fates and functions of transplanted mesenchymal stromal cells during bone repair. Stem Cell Reports 2022; 17:2318-2333. [PMID: 36150383 PMCID: PMC9561611 DOI: 10.1016/j.stemcr.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) transplantation could enhance bone repair. However, the cell fate of transplanted MSCs, in terms of their local distribution and spatial associations with other types of cells were poorly understood. Here, we developed a single-cell 3D spatial correlation (sc3DSC) method to track transplanted MSCs based on deep tissue microscopy of fluorescent nanoparticles (fNPs) and immunofluorescence of key proteins. Locally delivered fNP-labeled MSCs enhanced tibial defect repair, increased the number of stem cells and vascular maturity in mice. fNP-MSCs persisted in the defect throughout repair. But only a small portion of transplanted cells underwent osteogenic differentiation (OSX+); a significant portion has maintained their expression of mesenchymal stem cell and skeletal stem cell markers (SCA-1 and PRRX1). Our results contribute to the optimization of MSC-based therapies. The sc3DSC method may be useful in studying cell-based therapies for the regeneration of other tissue types or disease models. Transplanted marrow stromal cells associated with vessels during bone defect repair Small proportion of transplanted cells differentiated into osteogenic cells A proportion of transplanted cells maintained expressions of stem cell markers
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zeshun Li
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Runpeng Hou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Linhao Fu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Decheng Wu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Quanying Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kai Li
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
10
|
Holmes C, Ishida W, Perdomo-Pantoja A, Elder BD, Cottrill E, Locke J, Witham TF. Comparing the efficacy of adipose-derived and bone marrow-derived cells in a rat model of posterolateral lumbar fusion. J Orthop Res 2022; 40:909-916. [PMID: 34081344 DOI: 10.1002/jor.25111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 02/04/2023]
Abstract
Although bone marrow-derived mesenchymal stem cells (BMCs) have been widely used in spinal fusion procedures, adipose-derived stem cells (ASCs) offer a number of advantages as an alternative clinical cell source. This study directly compares the efficacy of ASCs and BMCs from the same donor animals to achieve successful fusion when combined with a clinical-grade bone graft substitute in a rat lumbar fusion model. ASCs and BMCs were isolated from the same Lewis donor rats and grown to passage 2 (P2). Single-level bilateral posterolateral intertransverse process lumbar fusion surgery was performed on syngeneic rats divided into three experimental groups: clinical-grade bone graft substitute alone (CBGS); CBGS+ rat ASCs (rASC); and, CBGS+ rat BMCs (rBMC). Eight weeks postoperatively, fusion was evaluated via micro-CT, manual palpation and histology. In vitro analysis of the osteogenic capacity of rBMCs and rASCs was also performed. Results indicated that the average fusion volume in the rASC group was the largest and was significantly larger than the CBGS group. Although the rASC group displayed the highest fusion rates via micro-CT and manual palpation, this difference was not statistically significant. Cell-seeded grafts showed more histological bone formation than cell-free grafts. P2 rASCs and rBMCs displayed similar in vitro osteogenic differentiation capacities. Overall, this study showed that, when combined with a clinical-grade bone graft substitute in a rat model, rASCs cells yielded the largest fusion masses and comparable fusion results to rBMCs. These results add to growing evidence that ASCs provide an attractive alternative to BMCs for spinal fusion procedures.
Collapse
Affiliation(s)
- Christina Holmes
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, Florida, USA
| | - Wataru Ishida
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Benjamin D Elder
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ethan Cottrill
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Locke
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Timothy F Witham
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Shen F, Shi Y. Recent Advances in Single-Cell View of Mesenchymal Stem Cell in Osteogenesis. Front Cell Dev Biol 2022; 9:809918. [PMID: 35071243 PMCID: PMC8766509 DOI: 10.3389/fcell.2021.809918] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoblasts continuously replenished by osteoblast progenitor cells form the basis of bone development, maintenance, and regeneration. Mesenchymal stem cells (MSCs) from various tissues can differentiate into the progenitor cell of osteogenic lineage and serve as the main source of osteoblasts. They also respond flexibly to regenerative and anabolic signals emitted by the surrounding microenvironment, thereby maintaining bone homeostasis and participating in bone remodeling. However, MSCs exhibit heterogeneity at multiple levels including different tissue sources and subpopulations which exhibit diversified gene expression and differentiation capacity, and surface markers used to predict cell differentiation potential remain to be further elucidated. The rapid advancement of lineage tracing methods and single-cell technology has made substantial progress in the characterization of osteogenic stem/progenitor cell populations in MSCs. Here, we reviewed the research progress of scRNA-seq technology in the identification of osteogenic markers and differentiation pathways, MSC-related new insights drawn from single-cell technology combined with experimental technology, and recent findings regarding the interaction between stem cell fate and niche in homeostasis and pathological process.
Collapse
Affiliation(s)
- Fangyuan Shen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Su Y, Chen X, Zhou H, Shaw S, Chen J, Isales CM, Zhao J, Shi X. Expression of long noncoding RNA Xist is induced by glucocorticoids. Front Endocrinol (Lausanne) 2022; 13:1005944. [PMID: 36187119 PMCID: PMC9516292 DOI: 10.3389/fendo.2022.1005944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory and immunosuppressive agents. However, their clinical usage is limited by severe multisystemic side effects. Glucocorticoid induced osteoporosis results in significant morbidity and mortality but the cellular and molecular mechanisms underlying GC-induced bone loss are not clear. GC use results in decreased osteoblast differentiation with increased marrow adiposity through effects on bone marrow stem cells. GC effects are transduced through its receptor (GR). To identify novel GR regulated genes, we performed RNA sequencing (RNA-Seq) analysis comparing conditional GR knockout mouse made by crossing the floxed GR animal with the Col I promoter-Cre, versus normal floxed GR without Cre, and that testing was specific for Col I promoter active cells, such as bone marrow mesenchymal stem/osteoprogenitor cells (MSCs) and osteoblasts. Results showed 15 upregulated genes (3- to 10-fold) and 70 downregulated genes (-2.7- to -10-fold), with the long noncoding RNA X-inactive specific transcript (Xist) downregulated the most. The differential expression of genes measured by RNA-Seq was validated by qRT-PCR analysis of selected genes and the GC/GR signaling-dependent expression of Xist was further demonstrated by GC (dexamethasone) treatment of GR-deficient MSCs in vitro and by GC injection of C57BL/6 mice (wild-type males and females) in vivo. Our data revealed that the long noncoding RNA Xist is a GR regulated gene and its expression is induced by GC both in vitro and in vivo. To our knowledge, this is the first evidence showing that Xist is transcriptionally regulated by GC/GR signaling.
Collapse
Affiliation(s)
- Yun Su
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Xing Chen
- Department of Mathematics, Logistical Engineering University, Chongqing, China
| | - Hongyan Zhou
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, United States
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Sean Shaw
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Jie Chen
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Augusta University, Augusta, GA, United States
| | - Carlos M. Isales
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, United States
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, United States
| | - Jing Zhao
- Institute of Interdisciplinary Complex Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xingming Shi
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, United States
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, United States
- *Correspondence: Xingming Shi,
| |
Collapse
|
13
|
Eisa NH, Sudharsan PT, Herrero SM, Herberg SA, Volkman BF, Aguilar-Pérez A, Kondrikov D, Elmansi AM, Reitman C, Shi X, Fulzele S, McGee-Lawrence ME, Isales CM, Hamrick MW, Johnson MH, Chen J, Hill WD. Age-associated changes in microRNAs affect the differentiation potential of human mesenchymal stem cells: Novel role of miR-29b-1-5p expression. Bone 2021; 153:116154. [PMID: 34403754 PMCID: PMC8935397 DOI: 10.1016/j.bone.2021.116154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/01/2021] [Accepted: 08/11/2021] [Indexed: 11/18/2022]
Abstract
Age-associated osteoporosis is widely accepted as involving the disruption of osteogenic stem cell populations and their functioning. Maintenance of the local bone marrow (BM) microenvironment is critical for regulating proliferation and differentiation of the multipotent BM mesenchymal stromal/stem cell (BMSC) population with age. The potential role of microRNAs (miRNAs) in modulating BMSCs and the BM microenvironment has recently gained attention. However, miRNAs expressed in rapidly isolated BMSCs that are naïve to the non-physiologic standard tissue culture conditions and reflect a more accurate in vivo profile have not yet been reported. Here we directly isolated CD271 positive (+) BMSCs within hours from human surgical BM aspirates without culturing and performed microarray analysis to identify the age-associated changes in BMSC miRNA expression. One hundred and two miRNAs showed differential expression with aging. Target prediction and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that the up-regulated miRNAs targeting genes in bone development pathways were considerably enriched. Among the differentially up-regulated miRNAs the novel passenger strand miR-29b-1-5p was abundantly expressed as a mature functional miRNA with aging. This suggests a critical arm-switching mechanism regulates the expression of the miR-29b-1-5p/3p pair shifting the normally degraded arm, miR-29b-1-5p, to be the dominantly expressed miRNA of the pair in aging. The normal guide strand miR-29b-1-3p is known to act as a pro-osteogenic miRNA. On the other hand, overexpression of the passenger strand miR-29b-1-5p in culture-expanded CD271+ BMSCs significantly down-regulated the expression of stromal cell-derived factor 1 (CXCL12)/ C-X-C chemokine receptor type 4 (SDF-1(CXCL12)/CXCR4) axis and other osteogenic genes including bone morphogenetic protein-2 (BMP-2) and runt-related transcription factor 2 (RUNX2). In contrast, blocking of miR-29b-1-5p function using an antagomir inhibitor up-regulated expression of BMP-2 and RUNX2 genes. Functional assays confirmed that miR-29b-1-5p negatively regulates BMSC osteogenesis in vitro. These novel findings provide evidence of a pathogenic anti-osteogenic role for miR-29b-1-5p and other miRNAs in age-related defects in osteogenesis and bone regeneration.
Collapse
Affiliation(s)
- Nada H Eisa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Periyasamy T Sudharsan
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, United States of America; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Sergio Mas Herrero
- Universitat de Barcelona, Unitat Farmacologia, Dpt. Fonaments Clínics, 08036 Barcelona, Spain
| | - Samuel A Herberg
- Departments of Ophthalmology and Visual Sciences, and Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, United States of America
| | - Brian F Volkman
- Biochemistry Department, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Alexandra Aguilar-Pérez
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Anatomy and Cell Biology, Indiana University School of Medicine in Indianapolis, IN, United States of America; Department of Cellular and Molecular Biology, School of Medicine, Universidad Central del Caribe, Bayamon 00956, Puerto Rico
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Charles Reitman
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America
| | - Xingming Shi
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Carlos M Isales
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Maribeth H Johnson
- Department of Population Health Sciences, Division of Biostatistics and Data Science Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Jie Chen
- Department of Population Health Sciences, Division of Biostatistics and Data Science Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.
| |
Collapse
|
14
|
Lin S, Pandruvada S, Yu H. Inhibition of Sphingosine-1-Phosphate Receptor 2 by JTE013 Promoted Osteogenesis by Increasing Vesicle Trafficking, Wnt/Ca 2+, and BMP/Smad Signaling. Int J Mol Sci 2021; 22:ijms222112060. [PMID: 34769490 PMCID: PMC8584480 DOI: 10.3390/ijms222112060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate receptor 2 (S1PR2) is a G protein-coupled receptor that regulates various immune responses. Herein, we determine the effects of a S1PR2 antagonist (JTE013) or a S1PR2 shRNA on osteogenesis by culturing murine bone marrow stromal cells (BMSCs) in osteogenic media with JTE013, dimethylsulfoxide (DMSO), a S1PR2 shRNA, or a control shRNA. Treatment with JTE013 or the S1PR2 shRNA increased alkaline phosphatase and alizarin red s staining, and enhanced alkaline phosphatase, RUNX2, osteocalcin, and osterix mRNA levels in BMSCs compared with the controls. Protein analysis revealed that a high dose of JTE013 (4 or 8 μM) increased vesicle trafficking-associated proteins (F-actin, clathrin, Early Endosome Antigen 1 (EEA1), and syntaxin 6) and Wnt/Ca2+ signaling. On the other hand, a low dose of JTE013 (1 to 2 μM) increased BMP/Smad signaling. In contrast, the S1PR2 shRNA reduced vesicle trafficking-associated proteins and attenuated Wnts and BMP/Smad signaling, but enhanced p-CaMKII compared with the control, suggesting that the S1PR2 shRNA influenced osteogenesis via different signaling pathways. Moreover, inhibiting protein trafficking by brefeldin A in BMSCs suppressed Wnts and BMPRs expressions. These data supported that enhanced osteogenesis in JTE013-treated BMSCs is associated with increased vesicle trafficking, which promotes the synthesis and transport of osteogenic protein and matrix vesicles and enhances matrix mineralization.
Collapse
|
15
|
Ma C, Gao J, Liang J, Dai W, Wang Z, Xia M, Chen T, Huang S, Na J, Xu L, Feng S, Dai K, Liu G. HDAC6 inactivates Runx2 promoter to block osteogenesis of bone marrow stromal cells in age-related bone loss of mice. Stem Cell Res Ther 2021; 12:484. [PMID: 34454588 PMCID: PMC8403388 DOI: 10.1186/s13287-021-02545-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/16/2021] [Indexed: 11/15/2022] Open
Abstract
Background Senile osteoporosis can cause bone fragility and increased risk for fractures and has been one of the most prevalent and severe diseases affecting the elderly population worldwidely. The underlying mechanisms are currently intensive areas of investigation. In age-related bone loss, decreased bone formation overweighs increased bone resorption. The molecular mechanisms underlying defective bone formation in age-related bone loss are not completely understood. In particular, the specific role of histone acetylation in age-related bone loss has not been examined thoroughly. Methods We employed 6- and 18-month-old mice to investigate the mechanisms of defective bone formation in age-related bone loss. Bone marrow stromal cells (BMSCs) were induced to undergo in vitro osteogenic differentiation. Chromatin immunoprecipitation (ChIP) was used to investigate the binding of histone deacetylases (HDACs) on Runx2 promoter in BMSCs. Luciferase reporter and transient transfection assay were employed to study Runx2 gene expression modulation by HDAC and androgen receptor (AR). siRNA and HDAC6 inhibitor, Tubastatin A, were used to inhibit HDAC6 in vitro. And systemic administration of Tubastatin A was used to block HDAC6 in vivo. Results Age-related trabecular bone loss was observed in 18-month-old mice compared with 6-month-old mice. In vitro osteogenic differentiation potential of BMSCs from 18-month-old mice was weaker than 6-month-old mice, in which there was Runx2 expression inactivation in BMSCs of 18-month-old mice compared with 6-month-old mice, which was attributable to HDAC6-mediated histone hypoacetylation in Runx2 promoter. There was competitive binding of HDAC6 and AR on Runx2 promoter to modulate Runx2 expression in BMSCs. More importantly, through siRNA- or specific inhibitor-mediated HDAC6 inhibition, we could activate Runx2 expression, rescue in vitro osteogenesis potential of BMSCs, and alleviate in vivo age-related bone loss of mice. Conclusion HDAC6 accumulation and histone hypoacetylation on Runx2 promoter contributed to the attenuation of in vitro osteogenic differentiation potential of BMSCs from aged mice. Through HDAC6 inhibition, we could activate Runx2 expression and osteogenic differentiation potential of BMSCs from aged mice and alleviate the age-related bone loss of aged mice. Our study will benefit not only for understanding the age-related bone loss, but also for finding new therapies to treat senile osteoporosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02545-w.
Collapse
Affiliation(s)
- Chao Ma
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Juan Gao
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Jun Liang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Weixiang Dai
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Zhenfei Wang
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Mengjiao Xia
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Tao Chen
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Sen Huang
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Jian Na
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Long Xu
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Shiming Feng
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Guangwang Liu
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, 221009, Jiangsu, China.
| |
Collapse
|
16
|
Eroglu B, Genova E, Zhang Q, Su Y, Shi X, Isales C, Eroglu A. Photobiomodulation has rejuvenating effects on aged bone marrow mesenchymal stem cells. Sci Rep 2021; 11:13067. [PMID: 34158600 PMCID: PMC8219765 DOI: 10.1038/s41598-021-92584-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/08/2021] [Indexed: 01/06/2023] Open
Abstract
The plasticity and proliferative capacity of stem cells decrease with aging, compromising their tissue regenerative potential and therapeutic applications. This decline is directly linked to mitochondrial dysfunction. Here, we present an effective strategy to reverse aging of mouse bone marrow mesenchymal stem cells (BM-MSCs) by restoring their mitochondrial functionality using photobiomodulation (PBM) therapy. Following the characterization of young and aged MSCs, our results show that a near-infrared PBM treatment delivering 3 J/cm2 is the most effective modality for improving mitochondrial functionality and aging markers. Furthermore, our results unveil that young and aged MSCs respond differently to the same modality of PBM: whereas the beneficial effect of a single PBM treatment dissipates within 7 h in aged stem cells, it is lasting in young ones. Nevertheless, by applying three consecutive treatments at 24-h intervals, we were able to obtain a lasting rejuvenating effect on aged MSCs. Our findings are of particular significance for improving autologous stem cell transplantation in older individuals who need such therapies most.
Collapse
Affiliation(s)
- Binnur Eroglu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
| | - Evan Genova
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
| | - Yun Su
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
| | - Xingming Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
| | - Carlos Isales
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Ali Eroglu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, CA-2004, Augusta, GA, 30912, USA.
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
17
|
Comparison of Freshly Isolated Adipose Tissue-derived Stromal Vascular Fraction and Bone Marrow Cells in a Posterolateral Lumbar Spinal Fusion Model. Spine (Phila Pa 1976) 2021; 46:631-637. [PMID: 32991510 DOI: 10.1097/brs.0000000000003709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Rat posterolateral lumbar fusion model. OBJECTIVE The aim of this study was to compare the efficacy of freshly isolated adipose tissue-derived stromal vascular fraction (A-SVF) and bone marrow cells (BMCs) cells in achieving spinal fusion in a rat model. SUMMARY OF BACKGROUND DATA Adipose tissue-derived stromal cells (ASCs) offer advantages as a clinical cell source compared to bone marrow-derived stromal cells (BMSCs), including larger available tissue volumes and reduced donor site morbidity. While pre-clinical studies have shown that ex vivo expanded ASCs can be successfully used in spinal fusion, the use of A-SVF cells better allows for clinical translation. METHODS A-SVF cells were isolated from the inguinal fat pads, whereas BMCs were isolated from the long bones of syngeneic 6- to 8-week-old Lewis rats and combined with Vitoss (Stryker) bone graft substitute for subsequent transplantation. Posterolateral spinal fusion surgery at L4-L5 was performed on 36 female Lewis rats divided into three experimental groups: Vitoss bone graft substitute only (VO group); Vitoss + 2.5 × 106 A-SVF cells/side; and, Vitoss + 2.5 × 106 BMCs/side. Fusion was assessed 8 weeks post-surgery via manual palpation, micro-computed tomography (μCT) imaging, and histology. RESULTS μCT imaging analyses revealed that fusion volumes and μCT fusion scores in the A-SVF group were significantly higher than in the VO group; however, they were not significantly different between the A-SVF group and the BMC group. The average manual palpation score was highest in the A-SVF group compared with the BMC and VO groups. Fusion masses arising from cell-seeded implants yielded better bone quality than nonseeded bone graft substitute. CONCLUSION In a rat model, A-SVF cells yielded a comparable fusion mass volume and radiographic rate of fusion to BMCs when combined with a clinical-grade bone graft substitute. These results suggest the feasibility of using freshly isolated A-SVF cells in spinal fusion procedures.Level of Evidence: N/A.
Collapse
|
18
|
Xu W, Ni C, Wang Y, Zheng G, Zhang J, Xu Y. Age-related trabecular bone loss is associated with a decline in serum Galectin-1 level. BMC Musculoskelet Disord 2021; 22:394. [PMID: 33906620 PMCID: PMC8080405 DOI: 10.1186/s12891-021-04272-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/16/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Senile osteoporosis with age-related bone loss is diagnosed depending on radiographic changes of bone and bone mineral density (BMD) measurement. However, radiographic alterations are usually signs of medium-late stage osteoporosis. Therefore, biomarkers have been proposed as indicators of bone loss. In the current study, Galectin-1 (Gal-1) showed age-related decline in mice serum. The role of Gal-1 in osteoporosis has not been investigated so far. Hence, the current study illustrated the relationship of serum Gal-1 level with bone loss. METHODS We employed 6- and 18-month-old mice to establish an animal model of age-related trabecular bone loss, whose bone density and microstructure were investigated by micro-CT. ELISA was used to measure the levels of Gal-1 in serum. The correlation analysis was performed to illustrate the relationship between serum Gal-1 levels and trabecular bone loss. In addition, immunohistochemistry was used to investigate the abundance of Gal-1 in bone marrow of mice. ELISA and western blot were performed to measure the secretion ability and protein expression of Gal-1 in bone marrow stromal cells (BMSC), hematopoietic stem cells (HSC) and myeloid progenitor (MP) respectively. Flow cytometry was used to measure BMSC number in bone marrow. Finally, male volunteers with age-related BMD decrease were recruited and the relationship between serum Gal-1 and BMD was analyzed. RESULTS Gal-1 showed age-related decline in mice serum. Serum Gal-1 was positively associated with BV/TV of femur, tibia and L1 vertebrae in mice. BMSC secreted more Gal-1 compared with HSC and MP. BMSC number in bone marrow was significantly lower in aged mice compared with young mice. Significant attenuation of Gal-1 protein expression was observed in BMSC and HSC from aged mice compared with young mice. Further, we found a decline in serum Gal-1 levels in men with age-related BMD decrease. There was positive correlation between BMD and serum Gal-1 levels in these men. CONCLUSIONS Age-related trabecular bone loss is associated with a decline in serum Gal-1 level in mice and men. Our study suggested Gal-1 had great potential to be a biomarker for discovering BMSC senescence, diagnosing early osteoporosis and monitoring trabecular bone loss.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.,Department of Orthopaedics, Shanghai Jiangong Hospital, Shanghai, 200083, China
| | - Cheng Ni
- Department of Orthopaedics, Shanghai Jiangong Hospital, Shanghai, 200083, China
| | - Yuxuan Wang
- Department of Orthopaedics, Shanghai Jiangong Hospital, Shanghai, 200083, China
| | - Guoqing Zheng
- Department of Orthopaedics, Shanghai Jiangong Hospital, Shanghai, 200083, China
| | - Jinshan Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
19
|
Li D, Liu J, Yang C, Tian Y, Yin C, Hu L, Chen Z, Zhao F, Zhang R, Lu A, Zhang G, Qian A. Targeting long noncoding RNA PMIF facilitates osteoprogenitor cells migrating to bone formation surface to promote bone formation during aging. Am J Cancer Res 2021; 11:5585-5604. [PMID: 33859765 PMCID: PMC8039942 DOI: 10.7150/thno.54477] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: The migration of mesenchymal osteoprogenitor cells (OPCs) to bone formation surface is the initial step of osteoblastogenesis before they undergo osteoblast differentiation and maturation for governing bone formation. However, whether the migration capacity of OPCs is compromised during aging and how it contributes to the aging-related bone formation reduction remain unexplored. In the present study, we identified a migration inhibitory factor (i.e., long noncoding RNA PMIF) and examined whether targeting lnc-PMIF could facilitate osteoprogenitor cells migrating to bone formation surface to promote bone formation during aging. Methods: Primary OPCs from young (6-momth-old) and aged (18-momth-old) C57BL/6 mice and stable lnc-PMIF knockdown/overexpression cell lines were used for in vitro and in vivo cell migration assay (i.e., wound healing assay, transwell assay and cell intratibial injection assay). RNA pulldown-MS/WB and RIP-qPCR were performed to identify the RNA binding proteins (RBPs) of lnc-PMIF. Truncations of lnc-PMIF and the identified RBP were engaged to determine the interaction motif between them by RNA pulldown-WB and EMSA. By cell-based therapy approach and by pharmacological approach, small interfering RNA (siRNA)-mediated lnc-PMIF knockdown were used in aged mice. The cell migration ability was evaluated by transwell assay and cell intratibial injection assay. The bone formation was evaluated by microCT analysis and bone morphometry analysis. Results: We reported that the decreased bone formation was accompanied by the reduced migration capacity of the bone marrow mesenchymal stem cells (BMSCs, the unique source of OPCs in bone marrow) in aged mice. We further identified that the long non-coding RNA PMIF (postulated migration inhibitory factor) (i.e., lnc-PMIF) was highly expressed in BMSCs from aged mice and responsible for the reduced migration capacity of aged OPCs to bone formation surface. Mechanistically, we found that lnc-PMIF could bind to human antigen R (HuR) for interrupting the HuR-β-actin mRNA interaction, therefore inhibit the expression of β-actin for suppressing the migration of aged OPCs. We also authenticated a functionally conserved human lncRNA ortholog of the murine lnc-PMIF. By cell-based therapy approach, we demonstrated that replenishing the aged BMSCs with small interfering RNA (siRNA)-mediated lnc-PMIF knockdown could promote bone formation in aged mice. By pharmacological approach, we showed that targeted delivery of lnc-PMIF siRNA approaching the OPCs around the bone formation surface could also promote bone formation in aged mice. Conclusion: Toward translational medicine, this study hints that targeting lnc-PMIF to facilitate aged OPCs migrating to bone formation surface could be a brand-new anabolic strategy for aging-related osteoporosis.
Collapse
|
20
|
Cao J, Ding K, Pan G, Rosario R, Su Y, Bao Y, Zhou H, Xu J, McGee Lawrence ME, Hamrick MW, Isales CM, Shi X. Deletion of PPARγ in Mesenchymal Lineage Cells Protects Against Aging-Induced Cortical Bone Loss in Mice. J Gerontol A Biol Sci Med Sci 2021; 75:826-834. [PMID: 32060555 DOI: 10.1093/gerona/glaa049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
Bone loss in aging is linked with chronic low-grade inflammation and the accumulation of marrowfat in animals and humans. Peroxisome proliferator-activated receptor gamma (PPARγ), an adipogenic regulator, plays key roles in these biological processes. However, studies of the roles of PPARγ in age-related bone loss and inflammation are lacking. We hypothesized that deletion of PPARγ in bone marrow mesenchymal lineage cells would reduce bone loss with aging, potentially through a reduction in fat-generated inflammatory responses and an increase in osteoblastic activity. In the present study, we show that mice deficient of PPARγ in Dermo1-expressing mesenchymal lineage cells (Dermo1-Cre:PPARγ fl/fl) have reduced fat mass and increased cortical bone thickness but that deficiency of PPARγ had limited effect on protection of trabecular bone with aging as demonstrated by dual-energy X-ray absorptiometry, µCT, and histomorphometric analyses. Conditional knockout of PPARγ reduced serum concentrations of adipokines, including adiponectin, resistin, and leptin, and reduced marrow stromal cell expression levels of inflammation-related genes. Inflammation genes involved in the interferon signaling pathway were reduced the most. These results demonstrate that disruption of the master adipogenic regulator, PPARγ, has a certain protective effect on aging-induced bone loss, suggesting that regulation of adipose function and modulation of interferon signaling are among the key mechanisms by which PPARγ regulates bone homeostasis during aging process.
Collapse
Affiliation(s)
- Jay Cao
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota
| | - Kehong Ding
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Guodong Pan
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Raysa Rosario
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Yun Su
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Yonggang Bao
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Hongyan Zhou
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Jianru Xu
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Meghan E McGee Lawrence
- Center for Healthy Aging, Augusta University, Georgia.,Department of Cell Biology and Anatomy, Augusta University, Georgia
| | - Mark W Hamrick
- Center for Healthy Aging, Augusta University, Georgia.,Department of Cell Biology and Anatomy, Augusta University, Georgia
| | - Carlos M Isales
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia.,Department of Medicine, Augusta University, Georgia
| | - Xingming Shi
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia.,Department of Medicine, Augusta University, Georgia
| |
Collapse
|
21
|
Cheng YH, Liu SF, Dong JC, Bian Q. Transcriptomic alterations underline aging of osteogenic bone marrow stromal cells. World J Stem Cells 2021; 13:128-138. [PMID: 33584984 PMCID: PMC7859986 DOI: 10.4252/wjsc.v13.i1.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/01/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Multipotent bone marrow stromal cells (BMSCs) are adult stem cells that form functional osteoblasts and play a critical role in bone remodeling. During aging, an increase in bone loss and reduction in structural integrity lead to osteoporosis and result in an increased risk of fracture. We examined age-dependent histological changes in murine vertebrae and uncovered that bone loss begins as early as the age of 1 mo. AIM To identify the functional alterations and transcriptomic dynamics of BMSCs during early bone loss. METHODS We collected BMSCs from mice at early to middle ages and compared their self-renewal and differentiation potential. Subsequently, we obtained the transcriptomic profiles of BMSCs at 1 mo, 3 mo, and 7 mo. RESULTS The colony-forming and osteogenic commitment capacity showed a comparable finding that decreased at the age of 1 mo. The transcriptomic analysis showed the enrichment of osteoblastic regulation genes at 1 mo and loss of osteogenic features at 3 mo. The BMSCs at 7 mo showed enrichment of adipogenic and DNA repair features. Moreover, we demonstrated that the WNT and MAPK signaling pathways were upregulated at 1 mo, followed by increased pro-inflammatory and apoptotic features. CONCLUSION Our study uncovered the cellular and molecular dynamics of bone aging in mice and demonstrated the contribution of BMSCs to the early stage of age-related bone loss.
Collapse
Affiliation(s)
- Yu-Hao Cheng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Shu-Fen Liu
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Jing-Cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qin Bian
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
22
|
Yi L, Ju Y, He Y, Yin X, Xu Y, Weng T. Intraperitoneal injection of Desferal® alleviated the age-related bone loss and senescence of bone marrow stromal cells in rats. Stem Cell Res Ther 2021; 12:45. [PMID: 33413663 PMCID: PMC7791659 DOI: 10.1186/s13287-020-02112-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022] Open
Abstract
Background Age-related bone loss plays a vital role in the development of osteoporosis and osteoporotic fracture. Bone marrow stromal cell (BMSC) senescence is highly associated with osteoporosis and limits the application of BMSCs in regenerative medicine. Hypoxia is an essential component for maintaining the normal physiology of BMSCs. We have reported that activation of hypoxia-induced factor by deletion of von Hippel-Lindau gene in osteochondral progenitor cells protected mice from aging-induced bone loss. However, whether pharmacologically manipulation of hypoxic niche would attenuate age-related bone loss and dysfunction of BMSCs is not well understood. Methods Twelve-month-old Sprague-Dawley rats were used as an aged model and were intraperitoneally injected with Desferal® (20, 60 mg/kg weight or vehicle), three times a week for a continuous 8-week period. Two-month-old young rats were set as a reference. After 8 weeks, micro-CT and HE staining were performed to determine the effect of Desferal® on bone loss. In order to investigate the effects of Desferal® on BMSC senescence, 12-month-old rats were treated with high-dose Desferal® (60 mg/kg weight) daily for 10 days. BMSCs were isolated and evaluated using CCK-8 assay, colony-forming cell assay, cell differentiation assay, laser confocal for reactive oxygen species (ROS) level, senescence-associated β-galactosidase (SA-β-gal) staining, and molecular expression test for stemness/senescence-associated genes. Results Micro-CT and HE staining showed that high-dose Desferal® significantly prevented bone loss in aged rats. Compared with vehicle group, the ex vivo experiments showed that short-term Desferal® administration could promote the potential of BMSC growth (proliferation and colony formation ability) and improve the rebalance of osteogenic and adipogenic differentiation, as well as rejuvenate senescent BMSCs (ROS level and SA-β-gal staining) and revise the expression of stemness/senescence-associated genes. The potential of BMSCs from 12M-H-Desferal® group at least partly revised to the level close to 2-month-old group. Conclusions The current study suggested that Desferal®, an iron-chelating agent, could alleviate age-related bone loss in middle-aged rats. Meanwhile, we found that short-term intraperitoneal injection of Desferal® partly rejuvenate BMSCs from aged rats. Overall, we demonstrated a novel role of Desferal® in rejuvenating aged BMSCs and preventing age-related bone loss. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02112-9.
Collapse
Affiliation(s)
- Lingxian Yi
- Department of Orthopaedics, Fourth medical center of PLA General Hospital, No. 51 Fucheng Road, Beijing, 10048, People's Republic of China.,Critical Care Medicine Department, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Yue Ju
- Department of Orthopaedics, Fourth medical center of PLA General Hospital, No. 51 Fucheng Road, Beijing, 10048, People's Republic of China.,Applied Biology Laboratory, Shenyang University of Chemical Technology, Shenyang, China
| | - Ying He
- Department of Orthopaedics, Fourth medical center of PLA General Hospital, No. 51 Fucheng Road, Beijing, 10048, People's Republic of China
| | - Xiushan Yin
- Applied Biology Laboratory, Shenyang University of Chemical Technology, Shenyang, China
| | - Ye Xu
- School of Mechanical Engineering and Automation, Beihang University, Beijing, People's Republic of China
| | - Tujun Weng
- Department of Orthopaedics, Fourth medical center of PLA General Hospital, No. 51 Fucheng Road, Beijing, 10048, People's Republic of China.
| |
Collapse
|
23
|
Aging of Bone Marrow Mesenchymal Stromal Cells: Hematopoiesis Disturbances and Potential Role in the Development of Hematologic Cancers. Cancers (Basel) 2020; 13:cancers13010068. [PMID: 33383723 PMCID: PMC7794884 DOI: 10.3390/cancers13010068] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary As for many other cancers, the risk of developing hematologic malignancies increases considerably as people age. In recent years, a growing number of studies have highlighted the influence of the aging microenvironment on hematopoiesis and tumor progression. Mesenchymal stromal cells are a major player in intercellular communication inside the bone marrow microenvironment involved in hematopoiesis support. With aging, their functions may be altered, leading to hematopoiesis disturbances which can lead to hematologic cancers. A good understanding of the mechanisms involved in mesenchymal stem cell aging and the consequences on hematopoiesis and tumor progression is therefore necessary for a better comprehension of hematologic malignancies and for the development of therapeutic approaches. Abstract Aging of bone marrow is a complex process that is involved in the development of many diseases, including hematologic cancers. The results obtained in this field of research, year after year, underline the important role of cross-talk between hematopoietic stem cells and their close environment. In bone marrow, mesenchymal stromal cells (MSCs) are a major player in cell-to-cell communication, presenting a wide range of functionalities, sometimes opposite, depending on the environmental conditions. Although these cells are actively studied for their therapeutic properties, their role in tumor progression remains unclear. One of the reasons for this is that the aging of MSCs has a direct impact on their behavior and on hematopoiesis. In addition, tumor progression is accompanied by dynamic remodeling of the bone marrow niche that may interfere with MSC functions. The present review presents the main features of MSC senescence in bone marrow and their implications in hematologic cancer progression.
Collapse
|
24
|
Eisa NH, Reddy SV, Elmansi AM, Kondrikova G, Kondrikov D, Shi XM, Novince CM, Hamrick MW, McGee-Lawrence ME, Isales CM, Fulzele S, Hill WD. Kynurenine Promotes RANKL-Induced Osteoclastogenesis In Vitro by Activating the Aryl Hydrocarbon Receptor Pathway. Int J Mol Sci 2020; 21:ijms21217931. [PMID: 33114603 PMCID: PMC7662708 DOI: 10.3390/ijms21217931] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/22/2020] [Indexed: 12/31/2022] Open
Abstract
There is increasing evidence of the involvement of the tryptophan metabolite kynurenine (KYN) in disrupting osteogenesis and contributing to aging-related bone loss. Here, we show that KYN has an effect on bone resorption by increasing osteoclastogenesis. We have previously reported that in vivo treatment with KYN significantly increased osteoclast number lining bone surfaces. Here, we report the direct effect of KYN on receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis in Raw 264.7 macrophage cells, and we propose a potential mechanism for these KYN-mediated effects. We show that KYN/RANKL treatment results in enhancement of RANKL-induced osteoclast differentiation. KYN drives upregulation and activation of the key osteoclast transcription factors, c-fos and NFATc1 resulting in an increase in the number of multinucleated TRAP+ osteoclasts, and in hydroxyapatite bone resorptive activity. Mechanistically, the KYN receptor, aryl hydrocarbon receptor (AhR), plays an important role in the induction of osteoclastogenesis. We show that blocking AhR signaling using an AhR antagonist, or AhR siRNA, downregulates the KYN/RANKL-mediated increase in c-fos and NFATc1 and inhibits the formation of multinucleated TRAP + osteoclasts. Altogether, this work highlights that the novelty of the KYN and AhR pathways might have a potential role in helping to regulate osteoclast function with age and supports pursuing additional research to determine if they are potential therapeutic targets for the prevention or treatment of osteoporosis.
Collapse
Affiliation(s)
- Nada H. Eisa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA; (N.H.E.); (A.M.E.); (G.K.); (D.K.)
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Sakamuri V. Reddy
- Darby Children’s Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Ahmed M. Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA; (N.H.E.); (A.M.E.); (G.K.); (D.K.)
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
| | - Galina Kondrikova
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA; (N.H.E.); (A.M.E.); (G.K.); (D.K.)
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA; (N.H.E.); (A.M.E.); (G.K.); (D.K.)
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
| | - Xing-Ming Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.W.H.); (M.E.M.-L.); (C.M.I.); (S.F.)
| | - Chad M. Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mark W. Hamrick
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.W.H.); (M.E.M.-L.); (C.M.I.); (S.F.)
- Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Meghan E. McGee-Lawrence
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.W.H.); (M.E.M.-L.); (C.M.I.); (S.F.)
- Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Carlos M. Isales
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.W.H.); (M.E.M.-L.); (C.M.I.); (S.F.)
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.W.H.); (M.E.M.-L.); (C.M.I.); (S.F.)
- Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - William D. Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA; (N.H.E.); (A.M.E.); (G.K.); (D.K.)
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
- Correspondence: ; Tel.: +1-(843)-792-6623
| |
Collapse
|
25
|
Anaya JM, Bollag WB, Hamrick MW, Isales CM. The Role of Tryptophan Metabolites in Musculoskeletal Stem Cell Aging. Int J Mol Sci 2020; 21:ijms21186670. [PMID: 32933099 PMCID: PMC7555967 DOI: 10.3390/ijms21186670] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022] Open
Abstract
Although aging is considered a normal process, there are cellular and molecular changes that occur with aging that may be detrimental to health. Osteoporosis is one of the most common age-related degenerative diseases, and its progression correlates with aging and decreased capacity for stem cell differentiation and proliferation in both men and women. Tryptophan metabolism through the kynurenine pathway appears to be a key factor in promoting bone-aging phenotypes, promoting bone breakdown and interfering with stem cell function and osteogenesis; however, little data is available on the impact of tryptophan metabolites downstream of kynurenine. Here we review available data on the impact of these tryptophan breakdown products on the body in general and, when available, the existing evidence of their impact on bone. A number of tryptophan metabolites (e.g., 3-hydroxykynurenine (3HKYN), kynurenic acid (KYNA) and anthranilic acid (AA)) have a detrimental effect on bone, decreasing bone mineral density (BMD) and increasing fracture risk. Other metabolites (e.g., 3-hydroxyAA, xanthurenic acid (XA), picolinic acid (PIA), quinolinic acid (QA), and NAD+) promote an increase in bone mineral density and are associated with lower fracture risk. Furthermore, the effects of other tryptophan breakdown products (e.g., serotonin) are complex, with either anabolic or catabolic actions on bone depending on their source. The mechanisms involved in the cellular actions of these tryptophan metabolites on bone are not yet fully known and will require further research as they are potential therapeutic targets. The current review is meant as a brief overview of existing English language literature on tryptophan and its metabolites and their effects on stem cells and musculoskeletal systems. The search terms used for a Medline database search were: kynurenine, mesenchymal stem cells, bone loss, tryptophan metabolism, aging, and oxidative stress.
Collapse
Affiliation(s)
- Jordan Marcano Anaya
- Universidad Central Del Caribe Laurel, Av. Sta. Juanita, Bayamón PR 00960, Puerto Rico;
| | - Wendy B. Bollag
- Department of Physiology, Augusta University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA;
| | - Mark W. Hamrick
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA;
| | - Carlos M. Isales
- Departments of Medicine, Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +706-721-0692
| |
Collapse
|
26
|
Elmansi AM, Hussein KA, Herrero SM, Periyasamy-Thandavan S, Aguilar-Pérez A, Kondrikova G, Kondrikov D, Eisa NH, Pierce JL, Kaiser H, Ding KH, Walker AL, Jiang X, Bollag WB, Elsalanty M, Zhong Q, Shi XM, Su Y, Johnson M, Hunter M, Reitman C, Volkman BF, Hamrick MW, Isales CM, Fulzele S, McGee-Lawrence ME, Hill WD. Age-related increase of kynurenine enhances miR29b-1-5p to decrease both CXCL12 signaling and the epigenetic enzyme Hdac3 in bone marrow stromal cells. Bone Rep 2020; 12:100270. [PMID: 32395570 PMCID: PMC7210406 DOI: 10.1016/j.bonr.2020.100270] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mechanisms leading to age-related reductions in bone formation and subsequent osteoporosis are still incompletely understood. We recently demonstrated that kynurenine (KYN), a tryptophan metabolite, accumulates in serum of aged mice and induces bone loss. Here, we report on novel mechanisms underlying KYN's detrimental effect on bone aging. We show that KYN is increased with aging in murine bone marrow mesenchymal stem cells (BMSCs). KYN reduces bone formation via modulating levels of CXCL12 and its receptors as well as histone deacetylase 3 (Hdac3). BMSCs responded to KYN by significantly decreasing mRNA expression levels of CXCL12 and its cognate receptors, CXCR4 and ACKR3, as well as downregulating osteogenic gene RUNX2 expression, resulting in a significant inhibition in BMSCs osteogenic differentiation. KYN's effects on these targets occur by increasing regulatory miRNAs that target osteogenesis, specifically miR29b-1-5p. Thus, KYN significantly upregulated the anti-osteogenic miRNA miR29b-1-5p in BMSCs, mimicking the up-regulation of miR-29b-1-5p in human and murine BMSCs with age. Direct inhibition of miR29b-1-5p by antagomirs rescued CXCL12 protein levels downregulated by KYN, while a miR29b-1-5p mimic further decreased CXCL12 levels. KYN also significantly downregulated mRNA levels of Hdac3, a target of miR-29b-1-5p, as well as its cofactor NCoR1. KYN is a ligand for the aryl hydrocarbon receptor (AhR). We hypothesized that AhR mediates KYN's effects in BMSCs. Indeed, AhR inhibitors (CH-223191 and 3',4'-dimethoxyflavone [DMF]) partially rescued secreted CXCL12 protein levels in BMSCs treated with KYN. Importantly, we found that treatment with CXCL12, or transfection with an miR29b-1-5p antagomir, downregulated the AhR mRNA level, while transfection with miR29b-1-5p mimic significantly upregulated its level. Further, CXCL12 treatment downregulated IDO, an enzyme responsible for generating KYN. Our findings reveal novel molecular pathways involved in KYN's age-associated effects in the bone microenvironment that may be useful translational targets for treating osteoporosis.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Khaled A Hussein
- Department of Oral Surgery and Medicine, National Research Centre, Cairo, Egypt
| | | | | | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology, Indiana University School of Medicine in Indianapolis, IN, United States of America.,Department of Cellular and Molecular Biology, School of Medicine, Universidad Central del Caribe, Bayamon 00956, Puerto Rico.,Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Galina Kondrikova
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Nada H Eisa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Jessica L Pierce
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Helen Kaiser
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Ke-Hong Ding
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Aisha L Walker
- Department of Medicine, Vascular Medicine Institute, University of Pittsburg School of Medicine, Pittsburg, PA 15261, United States of America
| | - Xue Jiang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wendy B Bollag
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, United States of America.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Mohammed Elsalanty
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Qing Zhong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Xing-Ming Shi
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Yun Su
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Population Health Science, Augusta University, Augusta, GA 30912, United States of America
| | - Monte Hunter
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America
| | - Charles Reitman
- Orthopaedics and Physical Medicine Department, Medical University of South Carolina, Charleston, SC 29403, United States of America
| | - Brian F Volkman
- Biochemistry Department, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Mark W Hamrick
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - Carlos M Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - Meghan E McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America.,Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, United States of America
| |
Collapse
|
27
|
Wang D, Cai G, Wang H, He J. TRAF3, a Target of MicroRNA-363-3p, Suppresses Senescence and Regulates the Balance Between Osteoblastic and Adipocytic Differentiation of Rat Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev 2020; 29:737-745. [PMID: 32111144 DOI: 10.1089/scd.2019.0276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential to differentiate into osteoblasts or adipocytes, and an imbalance between adipogenesis and osteogenesis causes age-related bone loss. In this study, we determined the influence of tumor necrosis factor receptor-associated factor 3 (TRAF3) on senescence and osteoblastic and adipocytic differentiation of rat BMSCs. TRAF3 expression increased during osteogenic differentiation but decreased during adipocytic differentiation of rat BMSCs, and compared with day 0 cultures, on day 14, the differences were significant. Overexpression of TRAF3 significantly promoted BMSC osteogenic differentiation and suppressed adipogenic differentiation and senescence. Furthermore, Traf3 was determined to be a target gene of miR-363-3p in BMSCs, and TRAF3 expression in BMSCs was reduced by miR-363-3p overexpression. This overexpression attenuated the effects of TRAF3 on BMSC adipogenic differentiation, osteogenic differentiation, and senescence. Taken together, these results uncovered the mechanism by which TRAF3 promotes BMSC osteogenic differentiation and suppresses adipogenic differentiation and senescence, indicating that the miR-363-3p-TRAF3 axis might be a novel therapeutic target for BMSC-based bone tissue engineering in osteoporosis.
Collapse
Affiliation(s)
- Dongliang Wang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiquan Cai
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiye He
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Stotesbury C, Alves-Peixoto P, Montoya B, Ferez M, Nair S, Snyder CM, Zhang S, Knudson CJ, Sigal LJ. α2β1 Integrin Is Required for Optimal NK Cell Proliferation during Viral Infection but Not for Acquisition of Effector Functions or NK Cell-Mediated Virus Control. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1582-1591. [PMID: 32015010 PMCID: PMC7065959 DOI: 10.4049/jimmunol.1900927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/05/2020] [Indexed: 01/13/2023]
Abstract
NK cells play an important role in antiviral resistance. The integrin α2, which dimerizes with integrin β1, distinguishes NK cells from innate lymphoid cells 1 and other leukocytes. Despite its use as an NK cell marker, little is known about the role of α2β1 in NK cell biology. In this study, we show that in mice α2β1 deficiency does not alter the balance of NK cell/ innate lymphoid cell 1 generation and slightly decreases the number of NK cells in the bone marrow and spleen without affecting NK cell maturation. NK cells deficient in α2β1 had no impairment at entering or distributing within the draining lymph node of ectromelia virus (ECTV)-infected mice or at becoming effectors but proliferated poorly in response to ECTV and did not increase in numbers following infection with mouse CMV (MCMV). Still, α2β1-deficient NK cells efficiently protected from lethal mousepox and controlled MCMV titers in the spleen. Thus, α2β1 is required for optimal NK cell proliferation but is dispensable for protection against ECTV and MCMV, two well-established models of viral infection in which NK cells are known to be important.
Collapse
Affiliation(s)
- Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Pedro Alves-Peixoto
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Savita Nair
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Shunchuan Zhang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Cory J Knudson
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
29
|
Yang BC, Kuang MJ, Kang JY, Zhao J, Ma JX, Ma XL. Human umbilical cord mesenchymal stem cell-derived exosomes act via the miR-1263/Mob1/Hippo signaling pathway to prevent apoptosis in disuse osteoporosis. Biochem Biophys Res Commun 2020; 524:883-889. [PMID: 32057365 DOI: 10.1016/j.bbrc.2020.02.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/01/2020] [Indexed: 12/29/2022]
Abstract
Disuse osteoporosis (DOP) is a common complication resulting from the lack of or disuse of mechanical loading and has been unsatisfactorily treated. We hypothesized that exosomes derived from human umbilical cord mesenchymal stem cells (HUCMSCs) could reduce bone marrow mesenchymal stem cell (BMSC) apoptosis in rat DOP via the miR-1263/Mob1/Hippo signaling pathway. To evaluate the function of exosomes derived from HUCMSCs (HUCMSC-Exos) in DOP, hind limb unloading (HLU)-induced DOP rat models were prepared. In vitro, the proliferation of BMSCs were evaluated using CCK-8 assays. Further, the apoptosis of BMSCs were evaluated using annexin V-FITC assay and Western blots. In vivo, the protective effects of HUCMSC-Exos were evaluated using HE staining and microCT analysis. The underlying molecular mechanism of exosome action on BMSC apoptosis through the miR-1263/Mob1/Hippo pathway was also investigated by high-throughput RNA sequencing, luciferase reporter assays, RNA-pull down assays and Western blots. The RNA-seq and q-PCR results showed that the level of miR-1263 was most abundant among differentially expressed microRNAs. Exosomal miR-1263 could bind to the 3'untranslated region (3' UTR) of Mob1 and exert its function by directly targeting Mob1 in recipient cells. The inhibition of Mob1 could activate YAP expression. Hippo inhibition reversed the in vitro HLU-induced apoptotic effect on BMSCs. The microCT and HE staining results indicated that HUCMSC-Exos ameliorated DOP in vivo. Exosomes derived from HUCMSCs are effective at inhibiting BMSC apoptosis and preventing rat DOP. This mechanism is mediated by the miR-1263/Mob1/Hippo signaling pathway.
Collapse
Affiliation(s)
- Bao-Cheng Yang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Ming-Jie Kuang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong University, Shandong, 250014, China
| | - Jia-Yu Kang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Jie Zhao
- Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, People's Republic of China
| | - Jian-Xiong Ma
- Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, People's Republic of China.
| | - Xin-Long Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
30
|
Khademi-Shirvan M, Ghorbaninejad M, Hosseini S, Baghaban Eslaminejad M. The Importance of Stem Cell Senescence in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:87-102. [PMID: 32026416 DOI: 10.1007/5584_2020_489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are an interesting tool in regenerative medicine and a unique cell-based therapy to treat aging-associated diseases. Successful MSC therapy needs a large-scale cell culture, and requires a prolonged in vitro cell culture that subsequently leads to cell senescence. Administration of senescent MSCs results in inefficient cell differentiation in the clinical setting. Therefore, it is of utmost importance to enhance our knowledge about the aging process and methods to detect cell senescence in order to overcome this challenge. Numerous studies have addressed senescence in various aspects. Here, we review the characteristics of MSCs, how aging affects their features, mechanisms involved in aging of MSCs, and potential approaches to detect MSC senescence in vitro.
Collapse
Affiliation(s)
- Maliheh Khademi-Shirvan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
31
|
Gilbert W, Bragg R, Elmansi AM, McGee-Lawrence ME, Isales CM, Hamrick MW, Hill WD, Fulzele S. Stromal cell-derived factor-1 (CXCL12) and its role in bone and muscle biology. Cytokine 2019; 123:154783. [PMID: 31336263 PMCID: PMC6948927 DOI: 10.1016/j.cyto.2019.154783] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
Musculoskeletal disorders are the leading cause of disability worldwide; two of the most prevalent of which are osteoporosis and sarcopenia. Each affect millions in the aging population across the world and the associated morbidity and mortality contributes to billions of dollars in annual healthcare cost. Thus, it is important to better understand the underlying pathologic mechanisms of the disease process. Regulatory chemokine, CXCL12, and its receptor, CXCR4, are recognized to be essential in the recruitment, localization, maintenance, development and differentiation of progenitor stem cells of the musculoskeletal system. CXCL12 signaling results in the development and functional ability of osteoblasts, osteoclasts, satellite cells and myoblasts critical to maintaining musculoskeletal homeostasis. Interestingly, one suggested pathologic mechanism of osteoporosis and sarcopenia is a decline in the regenerative capacity of musculoskeletal progenitor stem cells. Thus, because CXCL12 is critical to progenitor function, a disruption in the CXCL12 signaling axis might play a distinct role in these pathological processes. Therefore, in this article, we perform a review of CXCL12, its physiologic and pathologic function in bone and muscle, and potential targets for therapeutic development.
Collapse
Affiliation(s)
- William Gilbert
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States
| | - Robert Bragg
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States
| | - Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States
| | - Meghan E McGee-Lawrence
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States
| | - Mark W Hamrick
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
32
|
Liang M, Liu W, Peng Z, Lv S, Guan Y, An G, Zhang Y, Huang T, Wang Y. The therapeutic effect of secretome from human umbilical cord-derived mesenchymal stem cells in age-related osteoporosis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1357-1366. [PMID: 30977425 DOI: 10.1080/21691401.2019.1596945] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Senile osteoporosis is closely related to the loss of function of stem cells. In this study, we tried to investigate the potential of secretome from human umbilical cord-derived mesenchymal stem cells (hUCMSCs) in recovering stem cell ability from senescence and then delaying bone loss. We first harvested bone marrow-derived mesenchymal stem cells (BMSCs) from young and old rats and then compared their cellular characteristics such as cell growth, anti-senescence and differentiation. The results showed that these abilities were negatively affected by animal aging. Subsequently, aged BMSCs were exposed to secretome from hUCMSCs, and we found that this loss of cell potential can be modified by secretome treatment. Thereafter, the secretome was loaded into silk fibroin-based hydrogels and used for an in vivo animal study. The results showed that compared to the old untreated group, the bone formation capacity of aged rats was improved by local treatment of secretome-loaded silk fibroin hydrogels. In conclusion, these findings demonstrated that secretome from hUCMSCs has the capacity to recover stem cell potential and delay local bone loss in age-related osteoporosis, which could potentially be applied in osteoporosis therapy in the future.
Collapse
Affiliation(s)
- Min Liang
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Wanguo Liu
- b Department of Orthopaedic Surgery, China-Japan Union Hospital , Jilin University , Changchun , China
| | - Zhibin Peng
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Shihong Lv
- c Department of Gastroenterol , the Second Affiliated Hospital of Mudanjiang Medical University , Mudanjiang , China
| | - Ying Guan
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Gang An
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Yubo Zhang
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Tianwen Huang
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Yansong Wang
- a Department of Spine Surgery , the First Affiliated Hospital of Harbin Medical University , Harbin , China
| |
Collapse
|
33
|
Bollag AE, Guo T, Ding KH, Choudhary V, Chen X, Zhong Q, Xu J, Yu K, Awad ME, Elsalanty M, Johnson MH, McGee-Lawrence ME, Bollag WB, Isales CM. Monomethylfumarate protects against ovariectomy-related changes in body composition. J Endocrinol 2019; 243:JOE-18-0691.R3. [PMID: 31362266 PMCID: PMC6938560 DOI: 10.1530/joe-18-0691] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 07/30/2019] [Indexed: 12/31/2022]
Abstract
Osteoporosis, low bone mass that increases fracture susceptibility, affects approximately 75 million individuals in the United States, Europe and Japan, with the number of osteoporotic fractures expected to increase by more than 3-fold over the next 50 years. Bone mass declines with age, although the mechanisms for this decrease are unclear. Aging enhances production of reactive oxygen species, which can affect bone formation and breakdown. The multiple sclerosis drug Tecfidera contains dimethylfumarate, which is rapidly metabolized to monomethylfumarate (MMF); MMF is thought to function through nuclear factor erythroid-derived-2-like-2 (Nrf2), a transcription factor activated by oxidative stress which induces the expression of endogenous anti-oxidant systems. We hypothesized that MMF-elicited increases in anti-oxidants would inhibit osteopenia induced by ovariectomy, as a model of aging-related osteoporosis and high oxidative stress. We demonstrated that MMF activated Nrf2 and induced anti-oxidant Nrf2 target gene expression in bone marrow-derived mesenchymal stem cells. Sham-operated or ovariectomized adult female mice were fed chow with or without MMF and various parameters monitored. Ovariectomy produced the expected effects, decreasing bone mineral density and increasing body weight, fat mass, bone marrow adiposity and serum receptor activator of nuclear factor-kappa-B ligand (RANKL) levels. MMF decreased fat but not lean mass. MMF improved trabecular bone microarchitecture after adjustment for body weight, although the unadjusted data showed few differences; MMF also tended to increase adjusted cortical bone and to reduce bone marrow adiposity and serum RANKL levels. Because these results suggest the possibility that MMF might be beneficial for bone, further investigation seems warranted.
Collapse
Affiliation(s)
- Anna E. Bollag
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Tianyang Guo
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Ke-Hong Ding
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Vivek Choudhary
- Charlie Norwood VA Medical Center, Augusta, GA 30904
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Xunsheng Chen
- Charlie Norwood VA Medical Center, Augusta, GA 30904
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Qing Zhong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Jianru Xu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Mohamed E. Awad
- Department of Oral Biology, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Mohammed Elsalanty
- Department of Oral Biology, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Maribeth H. Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Meghan E. McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912
- Department of Orthopaedic Surgery, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Wendy B. Bollag
- Charlie Norwood VA Medical Center, Augusta, GA 30904
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Carlos M. Isales
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912
| |
Collapse
|
34
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|
35
|
Li X, Wu J, Liu S, Zhang K, Miao X, Li J, Shi Z, Gao Y. miR-384-5p Targets Gli2 and Negatively Regulates Age-Related Osteogenic Differentiation of Rat Bone Marrow Mesenchymal Stem Cells. Stem Cells Dev 2019; 28:791-798. [PMID: 30950325 DOI: 10.1089/scd.2019.0044] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aberrant microRNA expression correlates with age-related osteoporosis, which impairs bone formation by regulating osteoblastic activity, thus leading to age-related bone loss. In this study, we observed that miR-384-5p was significantly upregulated in bone marrow mesenchymal stem cells (BMSCs) from aged rats compared with BMSCs from young rats. In vitro functional assays revealed that overexpression of miR-384-5p in young BMSCs inhibited osteogenic differentiation and accelerated senescence, whereas knockdown of miR-384-5p in aged BMSCs had the opposite effects. Furthermore, we demonstrated that miR-384-5p inhibited the expression of Gli2 at both the mRNA and protein levels by directly binding to the 3' untranslated region of Gli2 mRNA. The osteogenic capacity of Gli2-knockdown BMSCs was rejuvenated by miR-384-5p inhibition. Finally, in vivo assays showed that the inhibition of miR-384-5p prevented bone loss and increased the osteogenic capacity in aged rats. Overall, our study suggests that miR-384-5p functions as a negative regulator of osteogenesis, indicating that the inhibition of miR-384-5p may be a therapeutic strategy against age-related bone loss.
Collapse
Affiliation(s)
- Xiaoming Li
- 1 Department of Orthopedic, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Jinhui Wu
- 2 Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Shu Liu
- 1 Department of Orthopedic, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Ke Zhang
- 2 Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Xiong Miao
- 1 Department of Orthopedic, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Jingfeng Li
- 1 Department of Orthopedic, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Zhicai Shi
- 1 Department of Orthopedic, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Yang Gao
- 3 Department of Orthopedics, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China
| |
Collapse
|
36
|
Fulzele S, Mendhe B, Khayrullin A, Johnson M, Kaiser H, Liu Y, Isales CM, Hamrick MW. Muscle-derived miR-34a increases with age in circulating extracellular vesicles and induces senescence of bone marrow stem cells. Aging (Albany NY) 2019; 11:1791-1803. [PMID: 30910993 PMCID: PMC6461183 DOI: 10.18632/aging.101874] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/10/2019] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are known to play important roles in cell-cell communication. Here we investigated the role of muscle-derived EVs and their microRNAs in the loss of bone stem cell populations with age. Aging in male and female C57BL6 mice was associated with a significant increase in expression of the senescence-associated microRNA miR-34a-5p (miR-34a) in skeletal muscle and in serum -derived EVs. Muscle-derived, alpha-sarcoglycan positive, EVs isolated from serum samples also showed a significant increase in miR-34a with age. EVs were isolated from conditioned medium of C2C12 mouse myoblasts and primary human myotubes after cells were treated with hydrogen peroxide to simulate oxidative stress. These EVs were shown to have elevated levels of miR-34a, and these EVs decreased viability of bone marrow mesenchymal (stromal) cells (BMSCs) and increased BMSC senescence. A lentiviral vector system was used to overexpress miR-34a in C2C12 cells, and EVs isolated from these transfected cells were observed to home to bone in vivo and to induce senescence and decrease Sirt1 expression of primary bone marrow cells ex vivo. These findings suggest that aged skeletal muscle is a potential source of circulating, senescence-associated EVs that may directly impact stem cell populations in tissues such as bone via their microRNA cargo.
Collapse
Affiliation(s)
- Sadanand Fulzele
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Bharati Mendhe
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Andrew Khayrullin
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Maribeth Johnson
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Helen Kaiser
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yutao Liu
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Carlos M. Isales
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mark W. Hamrick
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
37
|
Durham E, Howie RN, Warren G, LaRue A, Cray J. Direct Effects of Nicotine Exposure on Murine Calvaria and Calvarial Cells. Sci Rep 2019; 9:3805. [PMID: 30846819 PMCID: PMC6405741 DOI: 10.1038/s41598-019-40796-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/22/2019] [Indexed: 01/03/2023] Open
Abstract
Despite the link between adverse birth outcomes due to pre- and peri-natal nicotine exposure, research suggests 11% of US women continue to smoke or use alternative nicotine products throughout pregnancy. Maternal smoking has been linked to incidence of craniofacial anomalies. We hypothesized that pre-natal nicotine exposure may directly alter craniofacial development independent of the other effects of cigarette smoking. To test this hypothesis, we administered pregnant C57BL6 mice drinking water supplemented with 0, 50, 100 or 200 μg/ml nicotine throughout pregnancy. On postnatal day 15 pups were sacrificed and skulls underwent micro-computed tomography (µCT) and histological analyses. Specific nicotinic acetylcholine receptors, α3, α7, β2, β4 were identified within the calvarial growth sites (sutures) and centers (synchondroses). Exposing murine calvarial suture derived cells and isotype cells to relevant circulating nicotine levels alone and in combination with nicotinic receptor agonist and antagonists resulted in cell specific effects. Most notably, nicotine exposure increased proliferation in calvarial cells, an effect that was modified by receptor agonist and antagonist treatment. Currently it is unclear what component(s) of cigarette smoke is causative in birth defects, however these data indicate that nicotine alone is capable of disrupting growth and development of murine calvaria.
Collapse
Affiliation(s)
- Emily Durham
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - R Nicole Howie
- Department of Oral Health Sciences, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Graham Warren
- Departments of Radiation Oncology and Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Amanda LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 99 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - James Cray
- Department of Biomedical Education & Anatomy, The Ohio State University College of Medicine, 279 Hamilton Hall, 1645 Neil Ave, Columbus, Ohio, 43210, USA.
| |
Collapse
|
38
|
Fariyike B, Singleton Q, Hunter M, Hill WD, Isales CM, Hamrick MW, Fulzele S. Role of MicroRNA-141 in the Aging Musculoskeletal System: A Current Overview. Mech Ageing Dev 2019; 178:9-15. [PMID: 30528652 PMCID: PMC6998035 DOI: 10.1016/j.mad.2018.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/31/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
MicroRNA's are small non-coding RNAs that regulate the expression of genes by targeting the 3' UTR's of mRNA. Studies reveal that miRNAs play a pivotal role in normal musculoskeletal function such as mesenchymal stem cell differentiation, survivability and apoptosis, osteogenesis, and chondrogenesis. Changes in normal miRNA expression have been linked to a number of pathological disease processes. Additionally, with aging, it is noted that there is dysregulation in the normal function of stem cell differentiation, bone formation/degradation, chondrocyte function, and muscle degeneration. Due to the change in expression of miRNA in degenerative musculoskeletal pathology, it is believed that these molecules may be at least partially responsible for cellular dysfunction. A number of miRNAs have already been identified to play a role in osteoarthritis, osteoporosis and sarcopenia. One miRNA that has become of interest recently is miRNA 141. The purpose of this article is to review the current literature available on miRNA 141 and how it could play a role in osteoporosis, osteoarthritis and musculoskeletal pathology overall.
Collapse
Affiliation(s)
- Babatunde Fariyike
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Quante Singleton
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Monte Hunter
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - William D Hill
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Medicine, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States.
| |
Collapse
|
39
|
Khayrullin A, Krishnan P, Martinez-Nater L, Mendhe B, Fulzele S, Liu Y, Mattison JA, Hamrick MW. Very Long-Chain C24:1 Ceramide Is Increased in Serum Extracellular Vesicles with Aging and Can Induce Senescence in Bone-Derived Mesenchymal Stem Cells. Cells 2019; 8:cells8010037. [PMID: 30634626 PMCID: PMC6356348 DOI: 10.3390/cells8010037] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, function in cell-to-cell communication through delivery of proteins, lipids and microRNAs to target cells via endocytosis and membrane fusion. These vesicles are enriched in ceramide, a sphingolipid associated with the promotion of cell senescence and apoptosis. We investigated the ceramide profile of serum exosomes from young (24⁻40 yrs.) and older (75⁻90 yrs.) women and young (6⁻10 yrs.) and older (25⁻30 yrs.) rhesus macaques to define the role of circulating ceramides in the aging process. EVs were isolated using size-exclusion chromatography. Proteomic analysis was used to validate known exosome markers from Exocarta and nanoparticle tracking analysis used to characterize particle size and concentration. Specific ceramide species were identified with lipidomic analysis. Results show a significant increase in the average amount of C24:1 ceramide in EVs from older women (15.4 pmol/sample) compared to those from younger women (3.8 pmol/sample). Results were similar in non-human primate serum samples with increased amounts of C24:1 ceramide (9.3 pmol/sample) in older monkeys compared to the younger monkeys (1.8 pmol/sample). In vitro studies showed that primary bone-derived mesenchymal stem cells (BMSCs) readily endocytose serum EVs, and serum EVs loaded with C24:1 ceramide can induce BMSC senescence. Elevated ceramide levels have been associated with poor cardiovascular health and memory impairment in older adults. Our data suggest that circulating EVs carrying C24:1 ceramide may contribute directly to cell non-autonomous aging.
Collapse
Affiliation(s)
- Andrew Khayrullin
- Medical College of Georgia, Augusta University, CB1116 Laney Walker Blvd, Augusta, GA 30912, USA.
| | - Priyanka Krishnan
- Medical College of Georgia, Augusta University, CB1116 Laney Walker Blvd, Augusta, GA 30912, USA.
| | | | - Bharati Mendhe
- Medical College of Georgia, Augusta University, CB1116 Laney Walker Blvd, Augusta, GA 30912, USA.
| | - Sadanand Fulzele
- School of Medicine, Universidad Central Del Caribe, Bayamon, PR 00960, USA.
| | - Yutao Liu
- Medical College of Georgia, Augusta University, CB1116 Laney Walker Blvd, Augusta, GA 30912, USA.
| | - Julie A Mattison
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mark W Hamrick
- Medical College of Georgia, Augusta University, CB1116 Laney Walker Blvd, Augusta, GA 30912, USA.
| |
Collapse
|
40
|
Ding KH, Cain M, Davis M, Bergson C, McGee-Lawrence M, Perkins C, Hardigan T, Shi X, Zhong Q, Xu J, Bollag WB, Hill W, Elsalanty M, Hunter M, Isales MC, Lopez P, Hamrick M, Isales CM. Amino acids as signaling molecules modulating bone turnover. Bone 2018; 115:15-24. [PMID: 29499416 PMCID: PMC6110952 DOI: 10.1016/j.bone.2018.02.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/25/2018] [Accepted: 02/26/2018] [Indexed: 10/17/2022]
Abstract
Except for the essential amino acids (AAs), much of the focus on adequate dietary protein intake has been on total nitrogen and caloric intake rather than AA composition. Recent data, however, demonstrate that "amino-acid sensing" can occur through either intracellular or extracellular nutrient-sensing mechanisms. In particular, members of the class 3 G-protein coupled receptor family, like the calcium-sensing receptor are known to preferentially bind specific AAs, which then modulate receptor activation by calcium ions and thus potentially impact bone turnover. In pursuing the possibility of direct nutrient effects on bone cells, we examined individual AA effects on osteoprogenitor/bone marrow stromal cells (BMSCs), a key target for bone anabolism. We demonstrate that BMSCs express both intracellular and extracellular nutrient sensing pathways and that AAs are required for BMSC survival. In addition, certain AA types, like members of the aromatic AAs, can potently stimulate increases in intracellular calcium and ERK phosphorylation/activation. Further, based on the in vitro data, we examined the effect of specific AAs on bone mass. To better evaluate the impact of specific AAs, we added these to a low-protein diet. Our data demonstrate that a low-protein diet itself is associated with a significant drop in bone mineral density (BMD) in the older mice, related, at least in part, to an increase in osteoclastic activity. This drop in BMD in mice on the low-protein diet is prevented by addition of AAs from the aromatic group. Taken together our data show that AAs function as specific and selective signaling molecules in bone cells.
Collapse
Affiliation(s)
- Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Michael Cain
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA
| | - Michael Davis
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Clare Bergson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, USA
| | - Meghan McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA
| | - Crystal Perkins
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Trevor Hardigan
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Xingming Shi
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Wendy B Bollag
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Medicine, Medical College of Georgia, Augusta University, USA; Department of Physiology, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA; Charlie Norwood VA Medical Center, School of Dental Medicine, Augusta, GA 30912, USA
| | - William Hill
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA; Charlie Norwood VA Medical Center, School of Dental Medicine, Augusta, GA 30912, USA
| | - Mohammed Elsalanty
- Department of Oral Biology, School of Dental Medicine, Augusta, GA 30912, USA
| | - Monte Hunter
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA
| | - Maria C Isales
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Patricia Lopez
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Mark Hamrick
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Medicine, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA.
| |
Collapse
|
41
|
Howie RN, Herberg S, Durham E, Grey Z, Bennfors G, Elsalanty M, LaRue AC, Hill WD, Cray JJ. Selective serotonin re-uptake inhibitor sertraline inhibits bone healing in a calvarial defect model. Int J Oral Sci 2018; 10:25. [PMID: 30174329 PMCID: PMC6119683 DOI: 10.1038/s41368-018-0026-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 04/12/2018] [Accepted: 04/27/2018] [Indexed: 12/12/2022] Open
Abstract
Bone wound healing is a highly dynamic and precisely controlled process through which damaged bone undergoes repair and complete regeneration. External factors can alter this process, leading to delayed or failed bone wound healing. The findings of recent studies suggest that the use of selective serotonin reuptake inhibitors (SSRIs) can reduce bone mass, precipitate osteoporotic fractures and increase the rate of dental implant failure. With 10% of Americans prescribed antidepressants, the potential of SSRIs to impair bone healing may adversely affect millions of patients' ability to heal after sustaining trauma. Here, we investigate the effect of the SSRI sertraline on bone healing through pre-treatment with (10 mg·kg-1 sertraline in drinking water, n = 26) or without (control, n = 30) SSRI followed by the creation of a 5-mm calvarial defect. Animals were randomized into three surgical groups: (a) empty/sham, (b) implanted with a DermaMatrix scaffold soak-loaded with sterile PBS or (c) DermaMatrix soak-loaded with 542.5 ng BMP2. SSRI exposure continued until sacrifice in the exposed groups at 4 weeks after surgery. Sertraline exposure resulted in decreased bone healing with significant decreases in trabecular thickness, trabecular number and osteoclast dysfunction while significantly increasing mature collagen fiber formation. These findings indicate that sertraline exposure can impair bone wound healing through disruption of bone repair and regeneration while promoting or defaulting to scar formation within the defect site.
Collapse
Affiliation(s)
- R Nicole Howie
- Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Samuel Herberg
- Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Emily Durham
- Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Zachary Grey
- Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Grace Bennfors
- Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Mohammed Elsalanty
- Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
- Oral Biology, Augusta University, Augusta, GA, USA
- Orthopaedic Surgery, Augusta University, Augusta, GA, USA
- Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C LaRue
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA, USA
- Research Service of the Ralph H Johnson VA Medical Center, Charleston, SC, USA
| | - William D Hill
- Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
- Orthopaedic Surgery, Augusta University, Augusta, GA, USA
- Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
- Research Service of the Ralph H Johnson VA Medical Center, Charleston, SC, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - James J Cray
- Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, GA, USA.
- Department of Regenerative Medicine and Cellular Biology, Charleston, SC, USA.
- Division of Anatomy, College of Medicine, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
42
|
The positive effect of chick embryo and nutrient mixture on bone marrow- derived mesenchymal stem cells from aging rats. Sci Rep 2018; 8:7051. [PMID: 29728592 PMCID: PMC5935737 DOI: 10.1038/s41598-018-25563-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/24/2018] [Indexed: 12/18/2022] Open
Abstract
The aging of many mammalian tissues is associated with loss of functional adult stem cells, especially bone marrow-derived mesenchymal stem cells (BMSCs). This study was aimed to analyze the biological effect of chick embryo (CE) and nutrient mixture (NM) on the BMSCs of aging rats. The aging rat model was established to be induced by D-galactose (500 mg/kg/d) for 90 days. Meanwhile, aging rats were fed with CE and NM in different dose manner by intragastric administration. At the end of the experimental period, serum was collected from rats and used for BMSCs culture. Flow cytometric analysis was used to investigate the BMSCs surface markers. Alizarin Red and oil red O staining were performed to evaluate the multi-lineage differentiation of BMSCs. The results showed that CE plus NM increased the telomere length of BMSCs and promoted BMSCs proliferation. Moreover, CE plus NM administration promoted BMSCs differentiation into osteoblasts and suppressed differentiation into adipocytes. High-throughput sequencing analysis revealed that there were 326 genes were up-regulated and 59 genes were down-regulated in BMSCs of aging rats treated with CE plus NM. In conclusion, CE plus NM supplement had potential to delay aging through the recovery of BMSCs senescence and could be used as a safe effective approach for nutritional therapy of anti-aging.
Collapse
|
43
|
Bollag WB, Choudhary V, Zhong Q, Ding KH, Xu J, Elsayed R, Yu K, Su Y, Bailey LJ, Shi XM, Elsalanty M, Johnson MH, McGee-Lawrence ME, Isales CM. Deletion of protein kinase D1 in osteoprogenitor cells results in decreased osteogenesis in vitro and reduced bone mineral density in vivo. Mol Cell Endocrinol 2018; 461:22-31. [PMID: 28811183 PMCID: PMC5756499 DOI: 10.1016/j.mce.2017.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/14/2017] [Accepted: 08/10/2017] [Indexed: 01/08/2023]
Abstract
Protein kinase D1 (PRKD1) is thought to play a role in a number of cellular functions, including proliferation and differentiation. We hypothesized that PRKD1 in bone marrow-derived mesenchymal stem cells (BMMSC) could modulate osteogenesis. In BMMSCs from floxed PRKD1 mice, PRKD1 ablation with adenovirus-mediated Cre-recombinase expression inhibited BMMSC differentiation in vitro. In 3- and 6-month-old conditional knockout mice (cKO), in which PRKD1 was ablated in osteoprogenitor cells by osterix promoter-driven Cre-recombinase, bone mineral density (BMD) was significantly reduced compared with floxed control littermates. Microcomputed tomography analysis also demonstrated a decrease in trabecular thickness and bone volume fraction in cKO mice at these ages. Dynamic bone histomorphometry suggested a mineralization defect in the cKO mice. However, by 9 months of age, the bone appeared to compensate for the lack of PRKD1, and BMD was not different. Taken together, these results suggest a potentially important role for PRKD1 in bone formation.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Physiology, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Medicine, Augusta University, 30912, United States; Department of Oral Biology, Augusta University, 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, 30912, United States.
| | - Vivek Choudhary
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Department of Physiology, Augusta University, 30912, United States
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Ranya Elsayed
- Department of Oral Biology, Augusta University, 30912, United States
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Augusta University, 30912, United States
| | - Yun Su
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Lakiea J Bailey
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Xing-Ming Shi
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Mohammed Elsalanty
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Oral Biology, Augusta University, 30912, United States
| | - Maribeth H Johnson
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States; Department of Biostatistics and Epidemiology, Augusta University, 30912, United States
| | - Meghan E McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, 30912, United States
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| |
Collapse
|
44
|
Zhang W, Chen E, Chen M, Ye C, Qi Y, Ding Q, Li H, Xue D, Gao X, Pan Z. IGFBP7 regulates the osteogenic differentiation of bone marrow-derived mesenchymal stem cells via Wnt/β-catenin signaling pathway. FASEB J 2018; 32:2280-2291. [PMID: 29242275 DOI: 10.1096/fj.201700998rr] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor-binding protein 7 (IGFBP7), a low-affinity IGF binder, may play an important role in bone metabolism. However, its function in osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (BMSCs) remains unclear. Therefore, we investigated its effects on osteogenic differentiation. Overexpression of IGFBP7 enhanced the expression of osteo-specific genes and proteins, and IGFBP7 knockdown decreased osteogenesis-specific markers. More mineral deposits and higher alkaline phosphatase activity were observed after the up-regulation of IGFBP7. Moreover, β-catenin levels were up-regulated by the overexpression of IGFBP7 or the addition of extracellular IGFBP7 protein and were reduced by the depletion of IGFBP7. The increase in osteogenic differentiation due to the overexpression of IGFBP7 was partially decreased by specific Wnt/β-catenin signaling inhibitors. Using a rat tibial osteotomy model, a sheet of IGFBP7-overexpressing BMSCs improved bone healing, as demonstrated by imaging, biomechanical, and histologic analyses. Taken together, these findings indicate that IGFBP7 regulates the osteogenic differentiation of BMSCs partly via the Wnt/β-catenin signaling pathway.-Zhang, W., Chen, E., Chen, M., Ye, C., Qi, Y., Ding, Q., Li, H., Xue, D., Gao, X., Pan, Z. IGFBP7 regulates the osteogenic differentiation of bone marrow-derived mesenchymal stem cells via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nephrology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Yiying Qi
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Qianhai Ding
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Hang Li
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Xiang Gao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| |
Collapse
|
45
|
Irie K, Tomofuji T, Ekuni D, Fukuhara D, Uchida Y, Kataoka K, Kobayashi S, Kikuchi T, Mitani A, Shimazaki Y, Morita M. Age-related changes of CD4 + T cell migration and cytokine expression in germ-free and SPF mice periodontium. Arch Oral Biol 2017; 87:72-78. [PMID: 29274620 DOI: 10.1016/j.archoralbio.2017.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Increasing age is a potential risk factor for periodontal tissue breakdown, which may be affected by commensal flora. The aim of this study evaluated age-related changes in CD4+ T cells, C-C chemokine ligand 5 (CCL5), interleukin (IL)-17A, and receptor activator of nuclear factor-kappa B ligand (RANKL) expression using germ-free (GF) and conventionally reared (SPF) mice. DESIGN GF and SPF mice at 8 (n = 6/group) and 22 weeks old (n = 6/group) were used. Immunohistochemical analyses were performed to determine the effects of aging on protein expression in periodontal tissues. Age-related changes in alveolar bone were quantified using micro-CT analysis. RESULTS SPF mice, but not GF mice, showed an age-related increase in alveolar bone loss (P < 0.01). SPF mice at 22 weeks of age increased expression of CD4+ T cells, CCL5, IL-17A, and RANKL compared to those at 8 weeks of age in connective tissue and alveolar bone surface (P < 0.01). Furthermore, there was increased CD4+ T cells, which were co-expressed with IL-17A and RANKL in SPF mice at 22 weeks of age. On the other hand, the GF mice did not show any significant differences in CD4+ T cells, CCL5, IL-17A and RANKL expression between the two age groups. CONCLUSIONS SPF mice induced an age-related increase in CD4+ T cells co- expressed with IL-17A and RANKL, with occurring alveolar bone loss. In contrast, GF mice did not show age-related changes in CD4+ T cell migration and cytokine expression.
Collapse
Affiliation(s)
- Koichiro Irie
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Takaaki Tomofuji
- Department of Community Oral Health, Asahi University School of Dentistry, Gifu, Japan
| | - Daisuke Ekuni
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Daiki Fukuhara
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoko Uchida
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kota Kataoka
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuichiro Kobayashi
- Department of Periodontology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Takeshi Kikuchi
- Department of Periodontology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Akio Mitani
- Department of Periodontology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Yoshihiro Shimazaki
- Department of Preventive Dentistry and Dental Public Health, School of Dentistry, Aichi, Gakuin University, Nagoya, Japan
| | - Manabu Morita
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
46
|
El Refaey M, McGee-Lawrence ME, Fulzele S, Kennedy EJ, Bollag WB, Elsalanty M, Zhong Q, Ding KH, Bendzunas NG, Shi XM, Xu J, Hill WD, Johnson MH, Hunter M, Pierce JL, Yu K, Hamrick MW, Isales CM. Kynurenine, a Tryptophan Metabolite That Accumulates With Age, Induces Bone Loss. J Bone Miner Res 2017; 32:2182-2193. [PMID: 28727234 PMCID: PMC5685888 DOI: 10.1002/jbmr.3224] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/07/2017] [Accepted: 07/19/2017] [Indexed: 12/19/2022]
Abstract
Age-dependent bone loss occurs in humans and in several animal species, including rodents. The underlying causal mechanisms are probably multifactorial, although an age-associated increase in the generation of reactive oxygen species has been frequently implicated. We previously reported that aromatic amino acids function as antioxidants, are anabolic for bone, and that they may potentially play a protective role in an aging environment. We hypothesized that upon oxidation the aromatic amino acids would not only lose their anabolic effects but also potentially become a catabolic byproduct. When measured in vivo in C57BL/6 mice, the tryptophan oxidation product and kynurenine precursor, N-formylkynurenine (NFK), was found to increase with age. We tested the direct effects of feeding kynurenine (kyn) on bone mass and also tested the short-term effects of intraperitoneal kyn injection on bone turnover in CD-1 mice. μCT analyses showed kyn-induced bone loss. Levels of serum markers of osteoclastic activity (pyridinoline [PYD] and RANKL) increased significantly with kyn treatment. In addition, histological and histomorphometric studies showed an increase in osteoclastic activity in the kyn-treated groups in both dietary and injection-based studies. Further, kyn treatment significantly increased bone marrow adiposity, and BMSCs isolated from the kyn-injected mice exhibited decreased mRNA expression of Hdac3 and its cofactor NCoR1 and increased expression of lipid storage genes Cidec and Plin1. A similar pattern of gene expression is observed with aging. In summary, our data show that increasing kyn levels results in accelerated skeletal aging by impairing osteoblastic differentiation and increasing osteoclastic resorption. These data would suggest that kyn could play a role in age-induced bone loss. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mona El Refaey
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Meghan E. McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Eileen J. Kennedy
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA, 30602
| | - Wendy B. Bollag
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
- Department of Physiology, Augusta University, Augusta, Georgia, 30912
- Department of Oral Biology, Augusta University, Augusta, Georgia, 30912
- Department of Charlie Norwood VA Medical Center, Augusta, Georgia, 30912
| | - Mohammed Elsalanty
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Oral Biology, Augusta University, Augusta, Georgia, 30912
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Nathaniel G. Bendzunas
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA, 30602
| | - Xing-ming Shi
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - William D. Hill
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
- Department of Charlie Norwood VA Medical Center, Augusta, Georgia, 30912
| | - Maribeth H. Johnson
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, Georgia, 30912
| | - Monte Hunter
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Jessica L. Pierce
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Mark W. Hamrick
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Carlos M. Isales
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| |
Collapse
|
47
|
Davis C, Dukes A, Drewry M, Helwa I, Johnson MH, Isales CM, Hill WD, Liu Y, Shi X, Fulzele S, Hamrick MW. MicroRNA-183-5p Increases with Age in Bone-Derived Extracellular Vesicles, Suppresses Bone Marrow Stromal (Stem) Cell Proliferation, and Induces Stem Cell Senescence. Tissue Eng Part A 2017; 23:1231-1240. [PMID: 28363268 PMCID: PMC5689127 DOI: 10.1089/ten.tea.2016.0525] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 01/08/2023] Open
Abstract
Microvesicle- and exosome-mediated transport of microRNAs (miRNAs) represents a novel cellular and molecular pathway for cell-cell communication. In this study, we tested the hypothesis that these extracellular vesicles (EVs) and their miRNAs might change with age, contributing to age-related stem cell dysfunction. EVs were isolated from the bone marrow interstitial fluid (supernatant) of young (3-4 months) and aged (24-28 months) mice to determine whether the size, concentration, and miRNA profile of EVs were altered with age in vivo. Results show that EVs isolated from bone marrow are CD63 and CD9 positive, and the concentration and size distribution of bone marrow EVs are similar between the young and aged mice. Bioanalyzer data indicate that EVs from both young and aged mice are highly enriched in miRNAs, and the miRNA profile of bone marrow EVs differs significantly between the young and aged mice. Specifically, the miR-183 cluster (miR-96/-182/-183) is highly expressed in aged EVs. In vitro assays demonstrate that aged EVs are endocytosed by primary bone marrow stromal cells (BMSCs), and these aged EVs inhibit the osteogenic differentiation of young BMSCs. Transfection of BMSCs with miR-183-5p mimic reduces cell proliferation and osteogenic differentiation, increases senescence, and decreases protein levels of the miR-183-5p target heme oxygenase-1 (Hmox1). In vitro assays utilizing H2O2-induced oxidative stress show that H2O2 treatment of BMSCs increases the abundance of miR-183-5p in BMSC-derived EVs, and Amplex Red assays demonstrate that H2O2 is elevated in the bone marrow microenvironment with age. Together, these data indicate that aging and oxidative stress can significantly alter the miRNA cargo of EVs in the bone marrow microenvironment, which may in turn play a role in stem cell senescence and osteogenic differentiation by reducing Hmox1 activity.
Collapse
Affiliation(s)
- Colleen Davis
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Amy Dukes
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Michelle Drewry
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Inas Helwa
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Maribeth H Johnson
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Carlos M Isales
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - William D Hill
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Yutao Liu
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Xingming Shi
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Sadanand Fulzele
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Mark W Hamrick
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University , Augusta, Georgia
| |
Collapse
|
48
|
Strong AL, Jones RB, Glowacki J, Boue SM, Burow ME, Bunnell BA. Glycinol enhances osteogenic differentiation and attenuates the effects of age on mesenchymal stem cells. Regen Med 2017; 12:513-524. [PMID: 28718749 DOI: 10.2217/rme-2016-0148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM Phytoestrogens, such as glycinol, have recently gained significant attention as an alternative therapy for osteoporosis due to their structural similarity to estradiol and their bone-generating potential. METHODS The osteogenic effects of glycinol were investigated in human bone marrow mesenchymal stem cells (BMSCs) derived from older (>50 years old) and younger subjects (<25 years old). RESULTS BMSCs isolated from older donors demonstrated reduced osteogenesis. 17β-estradiol and glycinol exposure rescued the age-related reduction in osteogenic differentiation of BMSCs. These results correlated with the induction of osteogenic genes and estrogen receptor-α (ER-α) following glycinol treatment. ER antagonist studies further support that glycinol promotes osteogenesis through ER signaling. CONCLUSION The results from these studies support investigating glycinol as a potential preventive or treatment for osteoporosis.
Collapse
Affiliation(s)
- Amy L Strong
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Robert B Jones
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen M Boue
- Southern Regional Research Center, US Department of Agriculture, 1100 Robert E Lee Blvd, New Orleans, LA, USA
| | - Matthew E Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
49
|
Liu SY, He YB, Deng SY, Zhu WT, Xu SY, Ni GX. Exercise affects biological characteristics of mesenchymal stromal cells derived from bone marrow and adipose tissue. INTERNATIONAL ORTHOPAEDICS 2017; 41:1199-1209. [PMID: 28364139 DOI: 10.1007/s00264-017-3441-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/09/2017] [Indexed: 02/05/2023]
Abstract
Both bone marrow mesenchymal stromal cells (BMSCs) and adipose-derived mesenchymal stromal cells (ADSCs) are good sources for tissue engineering. To maximize therapeutic efficacy of MSCs, an appropriate source of MSCs should be selected according to their own inherent characteristics for future clinical application. Hence, this study was conducted to compare proliferative, differential and antiapoptosis abilities of both MSCs derived from exercised and sedentary rats under normal and hypoxia/serum deprivation conditions (H/SD). Our results showed that exercise may enhance proliferative ability and decrease adipogenic ability of BMSCs and ADSCs. However, positive effect of exercise on osteogenesis was only observed for BMSCs in either environment. Little effect was observed on the antiapoptotic ability of both MSC types. It was also suggested that biological characteristics of both types were partly changed. It is therefore believed that BMSCs derived from exercised rat on early passage may be a good cell source for bone tissue engineering.
Collapse
Affiliation(s)
- Sheng-Yao Liu
- Department of Orthopeadics and Traumatology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China
| | - Yong-Bin He
- Department of Orthopeadics and Traumatology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China
| | - Song-Yun Deng
- Department of Orthopeadics and Traumatology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China
| | - Wen-Ting Zhu
- Biomaterial Research Center, School of pharmaceutical sciences, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China
| | - Shao-Yong Xu
- Department of Orthopeadics and Traumatology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China
| | - Guo-Xin Ni
- Department of Orthopeadics and Traumatology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue (N), Guangzhou, 510515, China.
- Department of Rehabilitation Medicine, First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
| |
Collapse
|
50
|
Hamrick MW. Role of the Cytokine-like Hormone Leptin in Muscle-bone Crosstalk with Aging. J Bone Metab 2017; 24:1-8. [PMID: 28326295 PMCID: PMC5357607 DOI: 10.11005/jbm.2017.24.1.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
The cytokine-like hormone leptin is a classic adipokine that is secreted by adipocytes, increases with weight gain, and decreases with weight loss. Additional studies have, however, shown that leptin is also produced by skeletal muscle, and leptin receptors are abundant in both skeletal muscle and bone-derived mesenchymal (stromal) stem cells. These findings suggest that leptin may play an important role in muscle-bone crosstalk. Leptin treatment in vitro increases the expression of myogenic genes in primary myoblasts, and leptin treatment in vivo increases the expression of microRNAs involved in myogenesis. Bone marrow adipogenesis is associated with low bone mass in humans and rodents, and leptin can reduce marrow adipogenesis centrally through its receptors in the hypothalamus as well as directly via its receptors in bone marrow stem cells. Yet, central leptin resistance can increase with age, and low circulating levels of leptin have been observed among the frail elderly. Thus, aging appears to significantly alter leptin-mediated crosstalk among various organs and tissues. Aging is associated with bone loss and muscle atrophy, contributing to frailty, postural instability, and the incidence of falls. Therapeutic interventions such as protein and amino acid supplementation that can increase muscle mass and muscle-derived leptin may have multiple benefits for the elderly that can potentially reduce the incidence of falls and fractures.
Collapse
Affiliation(s)
- Mark W. Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|