1
|
DiVasta AD, Stamoulis C, Rubin CT, Gallagher JS, Kiel DP, Snyder BD, Gordon CM. Low-Magnitude Mechanical Signals to Preserve Skeletal Health in Female Adolescents With Anorexia Nervosa: A Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2441779. [PMID: 39480424 PMCID: PMC11528308 DOI: 10.1001/jamanetworkopen.2024.41779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 11/03/2024] Open
Abstract
Importance Malnourished adolescents and young adults with anorexia nervosa (AN) are at high risk for skeletal deficits. Objective To examine whether low-magnitude mechanical signals (LMMS) could preserve bone mineral density (BMD) throughout 6 months in adolescents and young adults with AN. Design, Setting, and Participants This double-blind, sham-controlled randomized clinical trial, conducted in a hospital-based specialty clinic, assessed female adolescents and young women without medical comorbidity or medication use that would compromise bone health. A total of 837 female adolescents were screened from January 1, 2012, to December 31, 2019, of whom 317 met the study criteria. Data analysis was performed from 2020 to 2024. Intervention Platform delivering low-magnitude mechanical signals (LMMS) (0.3 g at 32-37 Hz) or sham (ie, placebo) signals for 10 minutes daily for 6 months. Main Outcomes and Measures The primary outcome was trabecular volumetric BMD (vBMD) as measured by peripheral quantitative computed tomography of the tibia at baseline and 6 months. Secondary outcomes included cortical vBMD, cross-sectional area (CSA), areal BMD and body composition measured by dual-energy x-ray absorptiometry, and serum bone turnover markers. Results Forty female adolescents and young women (median [IQR] age, 16.3 [15.1-17.6] years; median [IQR] percentage median BMI for age, 87.2% [81.0%-91.6%]) completed the trial. Total bone vBMD changes were nonsignificant in both groups (95% CI for difference in median change between groups, -57.11 to 2.49): in the LMMS group, vBMD decreased from a median (IQR) of 313.4 (292.9-344.6) to 309.4 (290.4-334.0) mg/cm3, and in the placebo group, it increased from a median (IQR) of 308.5 (276.7-348.0) to 319.2 (309.9-338.4) mg/cm3. Total CSA at the 4% tibia site increased from a median (IQR) of 795.8 (695.0-844.8) mm2 to 827.5 (803.0-839.4) mm2 in the LMMS group, whereas in the placebo group, it decreased from 847.3 (770.5-915.3) mm2 to 843.3 (828.9-857.7) mm2 (95% CI for difference in median change between groups, 2.94-162.53). Median (IQR) trabecular CSA at the 4% tibia site increased from 616.3 (534.8-672.3) mm2 to 649.2 (638.0-661.4) mm2 in the LMMS group but decreased in the placebo group from 686.4 (589.0-740.0) mm2 to 647.9 (637.3-661.9) mm2 (95% CI for difference in median change between groups, 2.80-139.68 mm2). Changes in cortical vBMD, cortical section modulus, and muscle CSA were not significant between groups. The 6-month changes in trabecular and total bone CSA at the tibia 4% site (weight-bearing trabecular bone) were significantly different between groups (these measures increased in the LMMS group but decreased in the placebo group; total bone CSA: 95% CI, 2.94-162.53; P = .01; trabecular CSA: 95% CI, 2.80-139.68; P = .02). Greater increases in body mass index were seen in the placebo group (median [IQR] gain, 0.5 [-0.3 to +2.1]) than in the LMMS group (median [IQR] gain, +0.4 [-0.3 to +2.1]), perhaps due to differences in fat mass accrual. No adverse events occurred related to the LMMS intervention. Conclusions and Relevance In this randomized clinical trial of female adolescents and young women with AN, a 6-month LMMS intervention did not yield improvement in tibial trabecular vBMD. However, LMMS led to increases in total and trabecular CSA at the tibia. These results suggest an early positive response of increased bone turnover and trabecular bone quantity due to the LMMS intervention. Future studies should use a longer duration of intervention, consider strategies to optimize adherence, and potentially focus on a more profoundly malnourished patient population. Trial Registration ClinicalTrials.gov Identifier: NCT01100567.
Collapse
Affiliation(s)
- Amy D. DiVasta
- Division of Adolescent Medicine, Boston Children’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Catherine Stamoulis
- Division of Adolescent Medicine, Boston Children’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Clinton T. Rubin
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook
| | | | - Douglas P. Kiel
- Harvard Medical School, Boston, Massachusetts
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Brian D. Snyder
- Harvard Medical School, Boston, Massachusetts
- Department of Orthopedic Surgery, Boston Children’s Hospital, Boston, Massachusetts
| | - Catherine M. Gordon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| |
Collapse
|
2
|
Xiang YY, Won JH, Lee SJ, Baek KW. The Effect of Exercise on Mesenchymal Stem Cells and their Application in Obesity Treatment. Stem Cell Rev Rep 2024; 20:1732-1751. [PMID: 38954390 DOI: 10.1007/s12015-024-10755-x] [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] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
Mesenchymal stem cells (MSCs) have demonstrated considerable potential in tissue repair and the treatment of immune-related diseases, but there are problems with homing efficiency during MSCs transplantation. Exercise, as an intervention, has been shown to have an important impact on the properties of MSCs. This review summarizes the effects of exercise on the properties (including proliferation, apoptosis, differentiation, and homing) of bone marrow-derived MSCs and adipose-derived MSCs. Studies indicated that exercise enhances bone marrow-derived MSCs proliferation, osteogenic differentiation, and homing while reducing adipogenic differentiation. For adipose-derived MSCs, exercise enhances proliferation and reduces adipogenic differentiation. In addition, studies have investigated the therapeutic effects of combined therapy of MSCs transplantation with exercise on diseases of the bone, cardiac, and nervous systems. The combined therapy improves tissue repair by increasing the homing of transplanted MSCs and cytokine secretion (such as neurotrophin 4). Furthermore, MSCs transplantation also has potential for the treatment of obesity. Although the effect is not significant in weight loss, MSCs transplantation shows effects in controlling blood glucose, improving dyslipidemia, reducing inflammation, and improving liver disease. Finally, the potential role of combined MSCs transplantation and exercise therapy in addressing obesity is discussed.
Collapse
Affiliation(s)
- Ying-Ying Xiang
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea
| | - Jong-Hwa Won
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea
| | - Sam-Jun Lee
- Department of Sport Rehabilitation, College of Health, Tongmyong University, Welfare, and Education, Busan, 48520, Korea
| | - Kyung-Wan Baek
- Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Korea.
| |
Collapse
|
3
|
Wang XS, Chen Y, Zhao YW, Chen MW, Wang H. Assessing the association between a sedentary lifestyle and prevalence of primary osteoporosis: a community-based cross-sectional study among Chinese population. BMJ Open 2024; 14:e080243. [PMID: 38834324 PMCID: PMC11163664 DOI: 10.1136/bmjopen-2023-080243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/21/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVES To reveal the association between a sedentary lifestyle and the prevalence of primary osteoporosis (POP). DESIGN A community-based cross-sectional study was conducted. SETTING This study was conducted in communities in Hefei city, Anhui province, China. PARTICIPANTS A total of 1346 residents aged 40 and above underwent POP screening via calcaneus ultrasound bone mineral density (BMD) testing and completed a questionnaire survey. OUTCOME MEASURES The average daily sitting time was included in the study variable and used to assess sedentary behaviour. The 15 control variables included general information, dietary information and life behaviour information. Logistic regression was used to analyse the association between the POP prevalence and study or control variables in different models. RESULTS 1346 participants were finally included in the study. According to the 15 control variables, the crude model and 4 models were established. The analysis revealed that the average daily sitting time showed a significant correlation with the prevalence of POP in the crude model (OR=2.02, 95% CI=1.74 to 2.36, p<0.001), Model 1 (OR=2.65, 95% CI=2.21 to 3.17, p<0.001), Model 2 (OR=2.63, 95% CI=2.19 to 3.15, p<0.001), Model 3 (OR=2.62, 95% CI=2.18 to 3.15, p<0.001) and Model 4 (OR=2.58, 95% CI=2.14 to 3.11, p<0.001). Besides, gender, age and body mass index showed a significant correlation with the POP prevalence in all models. CONCLUSIONS This study suggests a potential association between a sedentary lifestyle and the prevalence of POP within the Chinese population. Modifying sedentary behaviours could contribute to a reduction in POP risk. However, longitudinal cohort studies are necessary to confirm this hypothesis in the future.
Collapse
Affiliation(s)
- Xiao-Song Wang
- Center for Big Data and Population Health of IHM, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Epidemiology and Biostatistics, Anhui Medical University, Hefei, Anhui, China
| | - Yong Chen
- Department of Social Medicine and Health Management, Anhui Medical University, Hefei, Anhui, China
| | - Yun-Wu Zhao
- Center for Big Data and Population Health of IHM, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ming-Wei Chen
- Center for Big Data and Population Health of IHM, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Heng Wang
- Center for Big Data and Population Health of IHM, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Epidemiology and Biostatistics, Anhui Medical University, Hefei, Anhui, China
- Department of Social Medicine and Health Management, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
4
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
5
|
Wang R, Wang Y, Niu Y, He D, Jin S, Li Z, Zhu L, Chen L, Wu X, Ding C, Wu T, Shi X, Zhang H, Li C, Wang X, Xie Z, Li W, Liu Y. Deep Learning-Predicted Dihydroartemisinin Rescues Osteoporosis by Maintaining Mesenchymal Stem Cell Stemness through Activating Histone 3 Lys 9 Acetylation. ACS CENTRAL SCIENCE 2023; 9:1927-1943. [PMID: 37901168 PMCID: PMC10604014 DOI: 10.1021/acscentsci.3c00794] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Indexed: 10/31/2023]
Abstract
Maintaining the stemness of bone marrow mesenchymal stem cells (BMMSCs) is crucial for bone homeostasis and regeneration. However, in vitro expansion and bone diseases impair BMMSC stemness, limiting its functionality in bone tissue engineering. Using a deep learning-based efficacy prediction system and bone tissue sequencing, we identify a natural small-molecule compound, dihydroartemisinin (DHA), that maintains BMMSC stemness and enhances bone regeneration. During long-term in vitro expansion, DHA preserves BMMSC stemness characteristics, including its self-renewal ability and unbiased differentiation. In an osteoporosis mouse model, oral administration of DHA restores the femur trabecular structure, bone density, and BMMSC stemness in situ. Mechanistically, DHA maintains BMMSC stemness by promoting histone 3 lysine 9 acetylation via GCN5 activation both in vivo and in vitro. Furthermore, the bone-targeted delivery of DHA by mesoporous silica nanoparticles improves its therapeutic efficacy in osteoporosis. Collectively, DHA could be a promising therapeutic agent for treating osteoporosis by maintaining BMMSC stemness.
Collapse
Affiliation(s)
- Ruoxi Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yu Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yuting Niu
- Central
Laboratory, National Center for Stomatology & National Clinical
Research Center for Oral Diseases & National Engineering Laboratory
for Digital and Material Technology of Stomatology & Beijing Key
Laboratory of Digital Stomatology & Research Center of Engineering
and Technology for Computerized Dentistry Ministry of Health &
NMPA Key Laboratory for Dental Materials & Translational Research
Center for Orocraniofacial Stem Cells and Systemic Health, Central
Laboratory, Peking University School and
Hospital for Stomatology, Beijing 100081, China
| | - Danqing He
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Shanshan Jin
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Zixin Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Lisha Zhu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Liyuan Chen
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xiaolan Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chengye Ding
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Tianhao Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xinmeng Shi
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - He Zhang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chang Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xin Wang
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Zhengwei Xie
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Weiran Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yan Liu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| |
Collapse
|
6
|
Yamada S, Ockermann PN, Schwarz T, Mustafa K, Hansmann J. Translation of biophysical environment in bone into dynamic cell culture under flow for bone tissue engineering. Comput Struct Biotechnol J 2023; 21:4395-4407. [PMID: 37711188 PMCID: PMC10498129 DOI: 10.1016/j.csbj.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Bone is a dynamic environment where osteocytes, osteoblasts, and mesenchymal stem/progenitor cells perceive mechanical cues and regulate bone metabolism accordingly. In particular, interstitial fluid flow in bone and bone marrow serves as a primary biophysical stimulus, which regulates the growth and fate of the cellular components of bone. The processes of mechano-sensory and -transduction towards bone formation have been well studied mainly in vivo as well as in two-dimensional (2D) dynamic cell culture platforms, which elucidated mechanically induced osteogenesis starting with anabolic responses, such as production of nitrogen oxide and prostaglandins followed by the activation of canonical Wnt signaling, upon mechanosensation. The knowledge has been now translated into regenerative medicine, particularly into the field of bone tissue engineering, where multipotent stem cells are combined with three-dimensional (3D) scaffolding biomaterials to produce transplantable constructs for bone regeneration. In the presence of 3D scaffolds, the importance of suitable dynamic cell culture platforms increases further not only to improve mass transfer inside the scaffolds but to provide appropriate biophysical cues to guide cell fate. In principle, the concept of dynamic cell culture platforms is rooted to bone mechanobiology. Therefore, this review primarily focuses on biophysical environment in bone and its translation into dynamic cell culture platforms commonly used for 2D and 3D cell expansion, including their advancement, challenges, and future perspectives. Additionally, it provides the literature review of recent empirical studies using 2D and 3D flow-based dynamic cell culture systems for bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Philipp Niklas Ockermann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Kamal Mustafa
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
- Department of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Germany
| |
Collapse
|
7
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
8
|
Pagnotti GM, Trivedi T, Wright LE, John SK, Murthy S, Pattyn RR, Willis MS, She Y, Suresh S, Thompson WR, Rubin CT, Mohammad KS, Guise TA. Low-Magnitude Mechanical Signals Combined with Zoledronic Acid Reduce Musculoskeletal Weakness and Adiposity in Estrogen-Deprived Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.12.531571. [PMID: 36993656 PMCID: PMC10054938 DOI: 10.1101/2023.03.12.531571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
UNLABELLED Combination treatment of Low-Intensity Vibration (LIV) with zoledronic acid (ZA) was hypothesized to preserve bone mass and muscle strength while reducing adipose tissue accrual associated with complete estrogen (E 2 )-deprivation in young and skeletally mature mice. Complete E 2 -deprivation (surgical-ovariectomy (OVX) and daily injection of aromatase inhibitor (AI) letrozole) were performed on 8-week-old C57BL/6 female mice for 4 weeks following commencement of LIV administration or control (no LIV), for 28 weeks. Additionally, 16-week-old C57BL/6 female E 2 -deprived mice were administered ±LIV twice daily and supplemented with ±ZA (2.5 ng/kg/week). By week 28, lean tissue mass quantified by dual-energy X-ray absorptiometry was increased in younger OVX/AI+LIV(y) mice, with increased myofiber cross-sectional area of quadratus femorii. Grip strength was greater in OVX/AI+LIV(y) mice than OVX/AI(y) mice. Fat mass remained lower in OVX/AI+LIV(y) mice throughout the experiment compared with OVX/AI(y) mice. OVX/AI+LIV(y) mice exhibited increased glucose tolerance and reduced leptin and free fatty acids than OVX/AI(y) mice. Trabecular bone volume fraction and connectivity density increased in the vertebrae of OVX/AI+LIV(y) mice compared to OVX/AI(y) mice; however, this effect was attenuated in the older cohort of E 2 -deprived mice, specifically in OVX/AI+ZA mice, requiring combined LIV with ZA to increase trabecular bone volume and strength. Similar improvements in cortical bone thickness and cross-sectional area of the femoral mid-diaphysis were observed in OVX/AI+LIV+ZA mice, resulting in greater fracture resistance. Our findings demonstrate that the combination of mechanical signals in the form of LIV and anti-resorptive therapy via ZA improve vertebral trabecular bone and femoral cortical bone, increase lean mass, and reduce adiposity in mice undergoing complete E 2 -deprivation. One Sentence Summary: Low-magnitude mechanical signals with zoledronic acid suppressed bone and muscle loss and adiposity in mice undergoing complete estrogen deprivation. TRANSLATIONAL RELEVANCE Postmenopausal patients with estrogen receptor-positive breast cancer treated with aromatase inhibitors to reduce tumor progression experience deleterious effects to bone and muscle subsequently develop muscle weakness, bone fragility, and adipose tissue accrual. Bisphosphonates (i.e., zoledronic acid) prescribed to inhibit osteoclast-mediated bone resorption are effective in preventing bone loss but may not address the non-skeletal effects of muscle weakness and fat accumulation that contribute to patient morbidity. Mechanical signals, typically delivered to the musculoskeletal system during exercise/physical activity, are integral for maintaining bone and muscle health; however, patients undergoing treatments for breast cancer often experience decreased physical activity which further accelerates musculoskeletal degeneration. Low-magnitude mechanical signals, in the form of low-intensity vibrations, generate dynamic loading forces similar to those derived from skeletal muscle contractility. As an adjuvant to existing treatment strategies, low-intensity vibrations may preserve or rescue diminished bone and muscle degraded by breast cancer treatment.
Collapse
|
9
|
Min S, Byeon Y, Kim M, Lee Y, Lee SH, Lee Y, Farooqi HMU, Lee HK, Paeng DG. Production enhancement of human adipose-derived mesenchymal stem cells by low-intensity ultrasound stimulation. Sci Rep 2022; 12:22041. [PMID: 36543825 PMCID: PMC9772213 DOI: 10.1038/s41598-022-24742-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Low-intensity ultrasound (LIUS) enhances the proliferation rate of various mammalian stem cells through mechanical stimulation. This study quantitively finds suitable LIUS stimulation parameters for increasing the proliferation rate of human adipose-derived mesenchymal stem cells (hAdMSCs) for mass production. Various stimulation conditions of LIUS were assessed based on the beam pattern of the ultrasonic transducer and the attenuation of the sound waves. Using optimal LIUS stimulation parameters for enhancing proliferation of hAdMSCs taken from bromodeoxyuridine (BrdU) incorporation assay, long-term culture of hAdMSCs was performed for 16 days. The resultant hAdMSCs were characterized for various biomarkers such as CD34-, CD45-, CD73+, CD95+, CD105+ and cytological staining and a cytokine array assay. LIUS stimulation parameters found for enhancing the hAdMSCs proliferation were the frequency of 5 MHz, an intensity of 300 mWcm-2, a duration of 10 min per day, and continuous waves with a 100% duty cycle. The LIUS stimulated hAdMSCs group showed a 3.25-fold increase in the cell number compared to the control group after 16 days of culture. By confirming the effects of quantitatively measured LIUS stimulation on the enhancement of hAdMSCs proliferation, this study may be a foundation for the applications of LIUS stimulation in the industrial-scale production of hAdMSCs.
Collapse
Affiliation(s)
- Soohong Min
- EHL Bio Inc, Gyeonggi, South Korea ,grid.411277.60000 0001 0725 5207Department of Ocean System Engineering, Jeju National University, Jeju, South Korea
| | | | - Min Kim
- EHL Bio Inc, Gyeonggi, South Korea
| | | | | | | | - Hafiz Muhammad Umer Farooqi
- grid.411277.60000 0001 0725 5207Department of Ocean System Engineering, Jeju National University, Jeju, South Korea
| | | | - Dong-Guk Paeng
- grid.411277.60000 0001 0725 5207Department of Ocean System Engineering, Jeju National University, Jeju, South Korea ,grid.27755.320000 0000 9136 933XDepartment of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA USA
| |
Collapse
|
10
|
Li X, Kordsmeier J, Nookaew I, Kim HN, Xiong J. Piezo1 stimulates mitochondrial function via cAMP signaling. FASEB J 2022; 36:e22519. [PMID: 36052712 PMCID: PMC10167693 DOI: 10.1096/fj.202200300r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/21/2022] [Accepted: 08/15/2022] [Indexed: 11/11/2022]
Abstract
Mechanical signals stimulate mitochondrial function but the molecular mechanisms are not clear. Here, we show that the mechanically sensitive ion channel Piezo1 plays a critical role in mitochondrial adaptation to mechanical stimulation. The activation of Piezo1 induced mitochondrial calcium uptake and oxidative phosphorylation (OXPHOS). In contrast, loss of Piezo1 reduced the mitochondrial oxygen consumption rate (OCR) and adenosine triphosphate (ATP) production in calvarial cells and these changes were associated with increased expression of the phosphodiesterases Pde4a and lower cyclic AMP (cAMP) levels. In addition, Piezo1 increased cAMP production and the activation of a cAMP-responsive transcriptional reporter. Consistent with this, cAMP was sufficient to increase mitochondrial OCR and the inhibition of phosphodiesterases augmented the increase in OCR induced by Piezo1. Moreover, the inhibition of cAMP production or activity of protein kinase A, a kinase activated by cAMP, prevented the increase in OCR induced by Piezo1. These results demonstrate that cAMP signaling contributes to the increase in mitochondrial OXPHOS induced by activation of Piezo1.
Collapse
Affiliation(s)
- Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jacob Kordsmeier
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Intawat Nookaew
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
11
|
Kim NR, David K, Sommers V, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Antonio L, Decallonne B, Claessens F, Vanderschueren D, Dubois V. Inactivation of AR or ERα in Extrahypothalamic Neurons Does not Affect Osteogenic Response to Loading in Male Mice. Endocrinology 2022; 163:6594680. [PMID: 35640239 DOI: 10.1210/endocr/bqac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 11/19/2022]
Abstract
Failure of bone mass maintenance in spite of functional loading is an important contributor to osteoporosis and related fractures. While the link between sex steroids and the osteogenic response to loading is well established, the underlying mechanisms are unknown, hampering clinical relevance. Androgens inhibit mechanoresponsiveness in male mice, but the cell type mediating this effect remains unidentified. To evaluate the role of neuronal sex steroid receptor signaling in the male bone's adaptive capacity, we subjected adult male mice with an extrahypothalamic neuron-specific knockout of the androgen receptor (N-ARKO) or the estrogen receptor alpha (N-ERαKO) to in vivo mechanical stimulation of the tibia. Loading increased cortical thickness in the control animals mainly through periosteal expansion, as total cross-sectional tissue area and cortical bone area but not medullary area were higher in the loaded than the unloaded tibia. Trabecular bone volume fraction also increased upon loading in the control group, mostly due to trabecular thickening. N-ARKO and N-ERαKO males displayed a loading response at both the cortical and trabecular bone compartments that was not different from their control littermates. In conclusion, we show that the presence of androgen receptor or estrogen receptor alpha in extrahypothalamic neurons is dispensable for the osteogenic response to mechanical loading in male mice.
Collapse
Affiliation(s)
- Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Vera Sommers
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204-5056, USA
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
- Basic and Translational Endocrinology, Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
12
|
Frequency-specific sensitivity of 3T3-L1 preadipocytes to low-intensity vibratory stimulus during adipogenesis. In Vitro Cell Dev Biol Anim 2022; 58:452-461. [PMID: 35713773 DOI: 10.1007/s11626-022-00696-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/20/2022] [Indexed: 11/05/2022]
Abstract
Adipocyte accumulation in the bone marrow is a severe complication leading to bone defects and reduced regenerative capacity. Application of external mechanical signals to bone marrow cellular niche is a non-invasive and non-pharmaceutical methodology to improve osteogenesis and suppress adipogenesis. However, in the literature, the specific parameters related to the nature of low-intensity vibratory (LIV) signals appear to be arbitrarily selected for amplitude, bouts, and applied frequency. In this study, we performed a LIV frequency sweep ranging from 30 to 120 Hz with increments of 15 Hz applied onto preadipocytes during adipogenesis for 10 d. We addressed the effect of LIV with different frequencies on single-cell density, adipogenic gene expression, lipid morphology, and triglycerides content. Results showed that LIV signals with 75-Hz frequency had the most significant suppressive effect during adipogenesis. Our results support the premise that mechanical-based interventions for suppressing adipogenesis may benefit from optimizing input parameters.
Collapse
|
13
|
Korkmaz HA, Özkan B. Impact of Obesity on Bone Metabolism in Children. J Pediatr Endocrinol Metab 2022; 35:557-565. [PMID: 35393850 DOI: 10.1515/jpem-2021-0714] [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: 11/26/2021] [Accepted: 02/28/2022] [Indexed: 11/15/2022]
Abstract
Obesity is an epidemic disease that can increase the incidence of type 2 diabetes, cardiovascular disease, malignancy, hypertension, and other health problems that affect the musculoskeletal system. There is a complex interaction between obesity and bone metabolism. In children with obesity, the peroxisome proliferator-activated receptor gamma pathway causes the differentiation of mesenchymal stem cells into adipocytes via osteoblasts, in which results in low bone mass and osteoporosis. Systemic inflammation in obesity has negative effects on bone metabolism. An increase in the number and size of adipose tissue and adipocytokines secreted from adipocytes affect the bone mass of the whole body with hormonal and biochemical effects. The skeletal effects of obesity are mediated by higher oxidative stress and increased production of proinflammatory cytokines. Osteoporosis due to obesity has increased morbidity and mortality in recent years, resulting in important health problems in developed and developing countries.
Collapse
Affiliation(s)
- Hüseyin Anıl Korkmaz
- Department of Pediatrics, Division of Pediatric Endocrinology, Dr Behcet Uz Child Disease and Surgery Training and Research Hospital, Izmir, Turkey
| | - Behzat Özkan
- Department of Pediatrics, Division of Pediatric Endocrinology, Dr Behcet Uz Child Disease and Surgery Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
14
|
BASKAN OZNUR, OZCIVICI ENGIN. VIABILITY OF 3T3-L1 PREADIPOCYTES IS MODULATED BY THE APPLIED FREQUENCY BUT NOT THE EXPOSURE DURATION OF LOW INTENSITY VIBRATORY STIMULATION. J MECH MED BIOL 2022. [DOI: 10.1142/s0219519422500063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the integral determinants in cell and tissue homeostasis and regeneration, and they can affect numerous biological process from proliferation to fate determination. Mechanical forces that possess low magnitude and high frequency characteristics are also known as low intensity vibrations (LIVs). These signals were studied widely on many cell types for regenerative purposes, however most of these studies select components of LIV signals (e.g., magnitude, frequency, duration, etc.) arbitrarily. Here, we addressed the effect of LIV applied frequency, LIV daily exposure time and fate induction on the viability of preadipocyte 3T3-L1 cells. For this, we performed a frequency sweep that was ranging from 30[Formula: see text]Hz to 120[Formula: see text]Hz with 15[Formula: see text]Hz increments applied for 5, 10 or 20[Formula: see text]min during quiescent growth or adipogenesis for up to 10 days. Results suggest that the applied frequency and fate induction was an important determinant of cell viability while daily exposure time had no effect. These findings contribute to the effort of optimizing a relevant mechanical stimulus that can inhibit adipogenesis.
Collapse
Affiliation(s)
- OZNUR BASKAN
- Department of Bioengineering, Izmir Institute of Technology Urla, Izmir 35430, Turkey
| | - ENGIN OZCIVICI
- Department of Bioengineering, Izmir Institute of Technology Urla, Izmir 35430, Turkey
| |
Collapse
|
15
|
CD34 +THY1 + synovial fibroblast subset in arthritic joints has high osteoblastic and chondrogenic potentials in vitro. Arthritis Res Ther 2022; 24:45. [PMID: 35168627 PMCID: PMC8845288 DOI: 10.1186/s13075-022-02736-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Synovial fibroblasts (SFs) in rheumatoid arthritis (RA) and osteoarthritis (OA) play biphasic roles in joint destruction and regeneration of bone/cartilage as mesenchymal stem cells (MSCs). Although MSCs contribute to joint homeostasis, such function is impaired in arthritic joints. We have identified functionally distinct three SF subsets characterized by the expression of CD34 and THY1 as follows: CD34+THY1+, CD34−THY1−, and CD34−THY1+. The objective of this study was to clarify the differentiation potentials as MSCs in each SF subset since both molecules would be associated with the MSC function. Methods SF subsets were isolated from synovial tissues of 70 patients (RA: 18, OA: 52). Expressions of surface markers associated with MSCs (THY1, CD34, CD73, CD271, CD54, CD44, and CD29) were evaluated in fleshly isolated SF subsets by flow cytometry. The differentiation potentials of osteogenesis, chondrogenesis, and adipogenesis were evaluated with histological staining and a quantitative polymerase chain reaction of differentiation marker genes. Small interfering RNA was examined to deplete THY1 in SFs. Results The expression levels of THY1+, CD73+, and CD271+ were highest and those of CD54+ and CD29+ were lowest in CD34+THY1+ among three subsets. Comparing three subsets, the calcified area, alkaline phosphatase (ALP)-stained area, and cartilage matrix subset were the largest in the CD34+THY1+ subset. Consistently, the expressions of differentiation markers of the osteoblasts (RUNX2, ALPL, and OCN) or chondrocytes (ACAN) were the highest in the CD34+THY1+ subset, indicating that the CD34+THY1+ subset possessed the highest osteogenic and chondrogenic potential among three subsets, while the differentiation potentials to adipocytes were comparable among the subsets regarding lipid droplet formations and the expression of LPL and PPARγ. The knockdown of THY1 in bulk SFs resulted in impaired osteoblast differentiation indicating some functional aspects in this stem-cell marker. Conclusion The CD34+THY1+ SF subset has high osteogenic and chondrogenic potentials. The preferential enhancement of MSC functions in the CD34+THY1+ subset may provide a new treatment strategy for regenerating damaged bone/cartilage in arthritic joints. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02736-7.
Collapse
|
16
|
Gao Q, Wang L, Wang S, Huang B, Jing Y, Su J. Bone Marrow Mesenchymal Stromal Cells: Identification, Classification, and Differentiation. Front Cell Dev Biol 2022; 9:787118. [PMID: 35047499 PMCID: PMC8762234 DOI: 10.3389/fcell.2021.787118] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (BMSCs), identified as pericytes comprising the hematopoietic niche, are a group of heterogeneous cells composed of multipotent stem cells, including osteochondral and adipocyte progenitors. Nevertheless, the identification and classification are still controversial, which limits their application. In recent years, by lineage tracing and single-cell sequencing, several new subgroups of BMSCs and their roles in normal physiological and pathological conditions have been clarified. Key regulators and mechanisms controlling the fate of BMSCs are being revealed. Cross-talk among subgroups of bone marrow mesenchymal cells has been demonstrated. In this review, we focus on recent advances in the identification and classification of BMSCs, which provides important implications for clinical applications.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Lipeng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China.,Wenzhou Institute of Shanghai University, Wenzhou, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
17
|
Liu X, Liu L, Zhao J, Wang H, Li Y. Mechanotransduction regulates inflammation responses of epicardial adipocytes in cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1080383. [PMID: 36589802 PMCID: PMC9800500 DOI: 10.3389/fendo.2022.1080383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is a crucial regulator in maintaining cardiovascular homeostasis by secreting various bioactive products to mediate the physiological function of the cardiovascular system. Accumulating evidence shows that adipose tissue disorders contribute to several kinds of cardiovascular disease (CVD). Furthermore, the adipose tissue would present various biological effects depending on its tissue localization and metabolic statuses, deciding the individual cardiometabolic risk. Crosstalk between adipose and myocardial tissue is involved in the pathophysiological process of arrhythmogenic right ventricular cardiomyopathy (ARVC), cardiac fibrosis, heart failure, and myocardial infarction/atherosclerosis. The abnormal distribution of adipose tissue in the heart might yield direct and/or indirect effects on cardiac function. Moreover, mechanical transduction is critical for adipocytes in differentiation, proliferation, functional maturity, and homeostasis maintenance. Therefore, understanding the features of mechanotransduction pathways in the cellular ontogeny of adipose tissue is vital for underlining the development of adipocytes involved in cardiovascular disorders, which would preliminarily contribute positive implications on a novel therapeutic invention for cardiovascular diseases. In this review, we aim to clarify the role of mechanical stress in cardiac adipocyte homeostasis and its interplay with maintaining cardiac function.
Collapse
Affiliation(s)
- Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfei Zhao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Hua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| |
Collapse
|
18
|
Li H, Luo H, Zhang Y, Liu L, Lin R. Association of Metabolic Dysfunction-Associated Fatty Liver Disease and Liver Stiffness With Bone Mineral Density in American Adults. Front Endocrinol (Lausanne) 2022; 13:891382. [PMID: 35846319 PMCID: PMC9280639 DOI: 10.3389/fendo.2022.891382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/24/2022] [Indexed: 12/23/2022] Open
Abstract
CONTEST The relationship between metabolic dysfunction-associated fatty liver disease (MAFLD) and liver stiffness and bone mineral density (BMD) remains unclear. OBJECTIVES We aimed to investigate the association between MAFLD and liver stiffness and BMD in the United States population. METHODS A cross-sectional study among 2031 participants over 50 years old in the National Health and Nutrition Examination Survey (NHANES) 2017-2018 was performed. All patients underwent vibration controlled transient elastography (VCTE) and dual-energy x-ray absorptiometry (DXA). The linear and logistic regression model were used to analyze the association between the MAFLD and liver stiffness and osteoporosis, with adjustments for known covariates. Furthermore, the sensitive analyses were conducted to explore the relationship between MAFLD and liver stiffness and whole osteoporosis (include femoral and lumbar osteoporosis). RESULTS MAFLD was prevalent in the study population, with a prevalence of 50.9% for men and 40.7% for women. The multiple linear models demonstrated positive associations between MAFLD and liver stiffness and total femur BMD, femur neck BMD, trochanter BMD, intertrochanter BMD. In multiple logistic regression models, both MAFLD and significant liver fibrosis were negatively associated with femoral osteoporosis (OR=0.41, 95% CI: 0.27 to 0.63; OR=0.67, 95% CI: 0.33-1.37, respectively). Nonetheless, when BMI was adjusted, the association between MAFLD and liver stiffness and osteoporosis became insignificant. Besides, as showed in the sensitive analyses, the relationship between MAFLD and liver stiffness and whole osteoporosis were stable. CONCLUSIONS These results suggest that MAFLD and liver stiffness were associated with higher femoral and lumbar bone mineral density in individuals aged over 50 years. But the results may be confounded by BMI.
Collapse
Affiliation(s)
- Hejun Li
- Department of Endocrinology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hengcong Luo
- Department of Endocrinology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Zhang
- Department of Endocrinology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lisi Liu
- Department of Nephrology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rong Lin
- Department of Endocrinology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Rong Lin,
| |
Collapse
|
19
|
Icariin and Icariside II Reciprocally Stimulate Osteogenesis and Inhibit Adipogenesis of Multipotential Stromal Cells through ERK Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8069930. [PMID: 34956384 PMCID: PMC8702327 DOI: 10.1155/2021/8069930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022]
Abstract
Herba Epimedii is a famous Chinese herbal medicine for treating bone diseases. Icariin and icariside II, the main chemical constituents, have attracted great attention from scientists for their potential as antiosteoporosis agents. Our study aimed to evaluate their effects on the lineage commitment of multipotential stromal cells (MSCs). The osteogenesis and adipogenesis of MSCs were assessed by ALP activity, calcium deposition, and adipocyte formation. The expression profiles and levels of osteogenic and adipogenic specific genes were evaluated by cDNA microarray and quantitative real-time PCR. The involvement of extracellular signal-regulated kinase (ERK) signaling was studied by enzyme-linked immunosorbent assay. Icariin and icariside II significantly increased ALP activity and mineralization during osteogenic differentiation of MSCs. Runx2, Col1, and Bmp2 were upregulated in the presence of icariin and icariside II. Meanwhile, they downregulated Pparg, Adipsin, and Cebpb expression during adipogenic differentiation. cDNA microarray revealed 57 differentially expressed genes during lineage commitment of MSCs. In addition, icariin and icariside II enhanced the phosphorylation of ERK, and the above biological effects were blocked by ERK inhibitor U0126. Icariin and icariside II may drive the final lineage commitment of MSCs towards osteogenesis and inhibit adipogenesis through the ERK signaling pathway. Both of them exert multiple osteoprotective effects and deserve more attention for their medicinal and healthcare prospects.
Collapse
|
20
|
Pagnotti GM, Thompson WR, Guise TA, Rubin CT. Suppression of cancer-associated bone loss through dynamic mechanical loading. Bone 2021; 150:115998. [PMID: 33971314 PMCID: PMC10044486 DOI: 10.1016/j.bone.2021.115998] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Patients afflicted with or being treated for cancer constitute a distinct and alarming subpopulation who exhibit elevated fracture risk and heightened susceptibility to developing secondary osteoporosis. Cancer cells uncouple the regulatory processes central for the adequate regulation of musculoskeletal tissue. Systemically taxing treatments to target tumors or disrupt the molecular elements driving tumor growth place considerable strain on recovery efforts. Skeletal tissue is inherently sensitive to mechanical forces, therefore attention to exercise and mechanical loading as non-pharmacological means to preserve bone during treatment and in post-treatment rehabilitative efforts have been topics of recent focus. This review discusses the dysregulation that cancers and the ensuing metabolic dysfunction that confer adverse effects on musculoskeletal tissues. Additionally, we describe foundational mechanotransduction pathways and the mechanisms by which they influence both musculoskeletal and cancerous cells. Functional and biological implications of mechanical loading at the tissue and cellular levels will be discussed, highlighting the current understanding in the field. Herein, in vitro, translational, and clinical data are summarized to consider the positive impact of exercise and low magnitude mechanical loading on tumor-bearing skeletal tissue.
Collapse
Affiliation(s)
- G M Pagnotti
- University of Texas - MD Anderson Cancer Center, Department of Endocrine, Neoplasia and Hormonal Disorders, Houston, TX, USA.
| | - W R Thompson
- Indiana University, Department of Physical Therapy, Indianapolis, IN, USA
| | - T A Guise
- University of Texas - MD Anderson Cancer Center, Department of Endocrine, Neoplasia and Hormonal Disorders, Houston, TX, USA
| | - C T Rubin
- Stony Brook University, Department of Biomedical Engineering, Stony Brook, NY, USA
| |
Collapse
|
21
|
Geng J, Zhang W, Chen C, Zhang H, Zhou A, Huang Y. Tracking the Differentiation Status of Human Neural Stem Cells through Label-Free Raman Spectroscopy and Machine Learning-Based Analysis. Anal Chem 2021; 93:10453-10461. [PMID: 34282890 DOI: 10.1021/acs.analchem.0c04941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability to noninvasively monitor stem cells' differentiation is important to stem cell studies. Raman spectroscopy is a non-harmful imaging approach that acquires the cellular biochemical signatures. Herein, we report the first use of label-free Raman spectroscopy to characterize the gradual change during the differentiation process of live human neural stem cells (NSCs) in the in vitro cultures. Raman spectra of 600-1800 cm-1 were measured with human NSC cultures from the undifferentiated stage (NSC-predominant) to the highly differentiated one (neuron-predominant) and subsequently analyzed using various mathematical methods. Hierarchical cluster analysis distinguished two cell types (NSCs and neurons) through the spectra. The subsequently derived differentiation rate matched that measured by immunocytochemistry. The key spectral biomarkers were identified by time-dependent trend analysis and principal component analysis. Furthermore, through machine learning-based analysis, a set of eight spectral data points were found to be highly accurate in classifying cell types and predicting the differentiation rate. The predictive accuracy was the highest using the artificial neural network (ANN) and slightly lowered using the logistic regression model and linear discriminant analysis. In conclusion, label-free Raman spectroscopy with the aid of machine learning analysis can provide the noninvasive classification of cell types at the single-cell level and thus accurately track the human NSC differentiation. A set of eight spectral data points combined with the ANN method were found to be the most efficient and accurate. Establishing this non-harmful and efficient strategy will shed light on the in vivo and clinical studies of NSCs.
Collapse
Affiliation(s)
- Junnan Geng
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| | - Wei Zhang
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| | - Cheng Chen
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| | - Han Zhang
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| | - Anhong Zhou
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| | - Yu Huang
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, ENGR 402, Logan, Utah 84322, United States
| |
Collapse
|
22
|
He L, Xiao J, Song L, Zhou R, Rong Z, He W, Dai F. HVEM Promotes the Osteogenesis of allo-MSCs by Inhibiting the Secretion of IL-17 and IFN-γ in Vγ4T Cells. Front Immunol 2021; 12:689269. [PMID: 34248977 PMCID: PMC8261146 DOI: 10.3389/fimmu.2021.689269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/02/2021] [Indexed: 12/29/2022] Open
Abstract
Bone defects are a common orthopaedic concern, and an increasing number of tissue-engineered bones (TEBs) are used to repair bone defects. Allogeneic mesenchymal stem cells (allo-MSCs) are used as seed cells in many approaches to develop TEB constructs, but the immune response caused by allogeneic transplantation may lead to transplant failure. V gamma 4 T (Vγ4T) cells play an important role in mediating the immune response in the early stage after transplantation; therefore, we wanted to verify whether suppressing Vγ4T cells by herpesvirus entry mediator (HVEM)/B and T lymphocyte attenuator (BTLA) signalling can promote MSCs osteogenesis in the transplanted area. In vitro experiments showed that the osteogenic differentiation of MSCs and Vγ4T cells was weakened after co-culture, and an increase in interleukin-17 (IL-17) and interferon-γ (IFN-γ) levels was detected in the culture supernatant. HVEM-transfected MSCs (MSCs-HVEM) still exhibited osteogenic differentiation activity after co-culture with Vγ4T cells, and the levels of IL-17 and IFN-γ in the co-culture supernatant were significantly reduced. In vivo experiments revealed that inflammation in the transplanted area was reduced and osteogenic repair was enhanced after Vγ4T cells were removed. MSCs-HVEM can also consistently contribute to reduced inflammation in the transplanted area and enhanced bone repair in wild-type (WT) mice. Therefore, our experiments verified that HVEM can promote the osteogenesis of allo-MSCs by inhibiting IL-17 and IFN-γ secretion from Vγ4T cells.
Collapse
Affiliation(s)
- Lei He
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jun Xiao
- Special Service Recuperation Center of Rocket Army, Guangzhou, China
| | - Lei Song
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Rui Zhou
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhigang Rong
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Weifeng He
- State Key Laboratory of Trauma, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Dai
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Zhang C, Wu J, Li X, Wang Z, Lu WW, Wong TM. Current Biological Strategies to Enhance Surgical Treatment for Rotator Cuff Repair. Front Bioeng Biotechnol 2021; 9:657584. [PMID: 34178957 PMCID: PMC8226184 DOI: 10.3389/fbioe.2021.657584] [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: 01/23/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Rotator cuff tear is one of the most common shoulder problems encountered by orthopedic surgeons. Due to the slow healing process and high retear rate, rotator cuff tear has distressed millions of people all around the world every year, especially for the elderly and active athletes. This disease significantly impairs patients' motor ability and reduces their quality of life. Besides conservative treatment, open and arthroscopic surgery contributes a lot to accelerate the healing process of rotator cuff tear. Currently, there are many emerging novel treatment methods to promote rotator cuff repair. A variety of biological stimulus has been utilized in clinical practice. Among them, platelet-rich plasma, growth factors, stem cells, and exosomes are the most popular biologics in laboratory research and clinical trials. This review will focus on the biologics of bioaugmentation methods for rotator cuff repair and tendon healing, including platelet-rich plasma, growth factors, exosomes and stem cells, etc. Relevant studies are summarized in this review and future research perspectives are introduced.
Collapse
Affiliation(s)
- Cheng Zhang
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jun Wu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiang Li
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zejin Wang
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Weijia William Lu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology (CAS), Shenzhen, China
| | - Tak-Man Wong
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
24
|
Michels MDL, Spivakoski CS, Réus BDS, Alves DMDS, Mattje PND, Hohl A, Ronsoni MF, Sande‐Lee S. Effect of whole body vibration on clinical and metabolic outcomes in adults with type 2 diabetes: an observational pilot trial. PRACTICAL DIABETES 2021. [DOI: 10.1002/pdi.2337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Manuella de L Michels
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Camila S Spivakoski
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Bruna da S Réus
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Débora M dos S Alves
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Priscila ND Mattje
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Alexandre Hohl
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Marcelo F Ronsoni
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| | - Simone Sande‐Lee
- Division of Endocrinology and Metabolism, University Hospital of the Federal University of Santa Catarina, Florianópolis SC Brazil
| |
Collapse
|
25
|
Mao L, Guo J, Hu L, Li L, Xu J, Zou J. The effects of biophysical stimulation on osteogenic differentiation and the mechanisms from ncRNAs. Cell Biochem Funct 2021; 39:727-739. [PMID: 34041775 DOI: 10.1002/cbf.3650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Ample proof showed that non-coding RNAs (ncRNAs) play a crucial role in proliferation and differentiation of osteoblasts and bone marrow stromal cells (BMSCs). Varied forms of biophysical stimuli like mechanical strain, fluid shear stress (FSS), microgravity and vibration are verified to regulate ncRNAs expression in osteogenic differentiation and influence the expression of target genes associated with osteogenic differentiation and ultimately regulate bone formation. The consequences of biophysical stimulation on osteogenic differentiation validate the prospect of exercise for the prevention and treatment of osteoporosis. In this review, we tend to summarize the studies on regulation of osteogenic differentiation by ncRNAs beneath biophysical stimulation and facilitate to reveal the regulatory mechanism of biophysical stimulation on ncRNAs, and provide an update for the prevention of bone metabolism diseases by exercise.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Linghui Hu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lexuan Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
26
|
Zhang W, Wang H, Yuan Z, Chu G, Sun H, Yu Z, Liang H, Liu T, Zhou F, Li B. Moderate mechanical stimulation rescues degenerative annulus fibrosus by suppressing caveolin-1 mediated pro-inflammatory signaling pathway. Int J Biol Sci 2021; 17:1395-1412. [PMID: 33867854 PMCID: PMC8040478 DOI: 10.7150/ijbs.57774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanical loading can induce or antagonize the extracellular matrix (ECM) synthesis, proliferation, migration, and inflammatory responses of annulus fibrosus cells (AFCs), depending on the loading mode and level. Caveolin-1 (Cav1), the core protein of caveolae, plays an important role in cellular mechanotransduction and inflammatory responses. In the present study, we presented that AFCs demonstrated different behaviors when subjected to cyclic tensile strain (CTS) for 24 h at a magnitude of 0%, 2%, 5% and 12%, respectively. It was found that 5% CTS had positive effects on cell proliferation, migration and anabolism, while 12% CTS had the opposite effects. Besides, cells exposed to interleukin-1β stimulus exhibited an increase expression in inflammatory genes, and the expression of these genes decreased after exposure to moderate mechanical loading with 5% CTS. In addition, 5% CTS decreased the level of Cav1 and integrin β1 and exhibited anti-inflammatory effects. Moreover, the expression of integrin β1 and p-p65 increased in AFCs transfected with Cav1 plasmids. In vivo results revealed that moderate mechanical stimulation could recover the water content and morphology of the discs. In conclusion, moderate mechanical stimulation restrained Cav1-mediated signaling pathway and exhibited anti-inflammatory effects on AFCs. Together with in vivo results, this study expounds the underlying molecular mechanisms on the effect of moderate mechanical stimulation on intervertebral discs (IVDs) and may provide a new therapeutic strategy for the treatment of IVD degeneration.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Zhangqin Yuan
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Heng Sun
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Zilin Yu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huan Liang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Tao Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Feng Zhou
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China.,China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Xia P, Wang X, Wang Q, Wang X, Lin Q, Cheng K, Li X. Low-Intensity Pulsed Ultrasound Promotes Autophagy-Mediated Migration of Mesenchymal Stem Cells and Cartilage Repair. Cell Transplant 2021; 30:963689720986142. [PMID: 33412895 PMCID: PMC7797574 DOI: 10.1177/0963689720986142] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cell (MSC) migration is promoted by low-intensity pulsed ultrasound (LIPUS), but its mechanism is unclear. Since autophagy is known to regulate cell migration, our study aimed to investigate if LIPUS promotes the migration of MSCs via autophagy regulation. We also aimed to investigate the effects of intra-articular injection of MSCs following LIPUS stimulation on osteoarthritis (OA) cartilage. For the in vitro study, rat bone marrow-derived MSCs were treated with an autophagy inhibitor or agonist, and then they were stimulated by LIPUS. Migration of MSCs was detected by transwell migration assays, and stromal cell-derived factor-1 (SDF-1) and C-X-C chemokine receptor type 4 (CXCR4) protein levels were quantified. For the in vivo study, a rat knee OA model was generated and treated with LIPUS after an intra-articular injection of MSCs with autophagy inhibitor added. The cartilage repair was assessed by histopathological analysis and extracellular matrix protein expression. The in vitro results suggest that LIPUS increased the expression of SDF-1 and CXCR4, and it promoted MSC migration. These effects were inhibited and enhanced by autophagy inhibitor and agonist, respectively. The in vivo results demonstrate that LIPUS significantly enhanced the cartilage repair effects of MSCs on OA, but these effects were blocked by autophagy inhibitor. Our results suggest that the migration of MSCs was enhanced by LIPUS through the activation autophagy, and LIPUS improved the protective effect of MSCs on OA cartilage via autophagy regulation.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Xinwei Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Qi Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
| |
Collapse
|
28
|
Fan Z, Chen Z, Zhang H, Nie Y, Xu S. Gradient Mineralized and Porous Double-Network Hydrogel Effectively Induce the Differentiation of BMSCs into Osteochondral Tissue In Vitro for Potential Application in Cartilage Repair. Macromol Biosci 2020; 21:e2000323. [PMID: 33356012 DOI: 10.1002/mabi.202000323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/02/2020] [Indexed: 02/06/2023]
Abstract
At present, it is a considerable challenge to mimic the complex architecture of osteochondral (OC) tissue. In this study, a porous and gradient mineralized double-network hydrogel is synthesized and used to induce bone marrow mesenchymal stem cells (BMSCs) to differentiate into the desired OC tissue depending only on the material and mechanical properties. Physical and chemical characterizations show that hydroxyapatite nanoparticles grow and fill into the pores of the hydrogel, and their content presents a gradient change in different layers of hydrogel. The synthesized hydrogel has excellent mechanical properties and the compression strength with different mineralization degrees varies from 27 to 380 kPa, which fully meets the needs of increased mechanical strength of articular cartilage from the surface to the deep layer. Besides, the synthesized hydrogel has good biocompatibility that can promote the proliferation and growth of BMSCs. More importantly, the results of histochemistry, immunohistochemistry, and real time polymerase chain reaction show that gradient mineralized hydrogel can induce BMSCs to differentiate into the desired chondrocytes and osteoblasts in different layers of hydrogels, indicating that OC tissues can be successfully constructed through a simple induction differentiation of gradient mineralized hydrogel.
Collapse
Affiliation(s)
- Zengjie Fan
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Zizi Chen
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Hui Zhang
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| | - Yingying Nie
- Institute of Sensing Technology, Gansu Academy of Sciences, Lanzhou, Gansu, 730000, P. R. China
| | - Shumei Xu
- Department of General Surgery, the 940th Hospital of Joint Logistics Support Force, PLA, Lanzhou, Gansu, 730050, P. R. China
| |
Collapse
|
29
|
Moussi K, Abusamra DB, Yassine O, Merzaban J, Kosel J. Strain-induced Differentiation of Mesenchymal Stem Cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2239-2244. [PMID: 33018453 DOI: 10.1109/embc44109.2020.9176273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Directing the fate of human mesenchymal stem/stromal cells (hMSCs) toward bone formation using mechanical strain is a promising approach in regenerative medicine related to bone diseases. Numerous studies have evaluated the effects of vibration or cyclic tensile strain on MSCs towards developing a mechanically-based method for stimulating differentiation. Here, we study the differentiation of hMSCs cultured on elastic polydimethylsiloxane (PDMS) membrane, which is magnetically actuated to induce periodically varying strain. The strain distribution across the membrane was calculated by finite-element modeling and demonstrates three main areas of different strain amplitudes. The strain effect on the hMSCs was evaluated by measuring the mineralization of differentiated hMSCs using Alizarin S red stain. The results indicate a strain-dependent differentiation of hMSCs, where the highest region of strain on the membrane resulted in the most accelerated differentiation. Osteogenic differentiation was achieved as early as two weeks, which is significantly sooner than control hMSCs treated with osteogenic media alone.
Collapse
|
30
|
Zhou H, Trudel G, Alexeev K, Laneuville O. Reversibility of marrow adipose accumulation and reduction of trabecular bone in the epiphysis of the proximal tibia. Acta Histochem 2020; 122:151604. [PMID: 33066832 DOI: 10.1016/j.acthis.2020.151604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/28/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Mechanical stimuli play an important role in the homeostasis of trabecular bone and marrow adipose tissue, particularly for the weight-bearing skeleton. Prolonged immobilization and disuse have been shown to reduce trabecular bone content and increase marrow adipose tissue in the bones of lower limb joints such as the knee. However, details on the temporal response of this relationship to prolonged immobilization and its reversibility is limited. Forty rats had one knee immobilized at 45° of flexion for 2, 4, 8, or 16 weeks and subsequently remobilized for 0 or 8 weeks. The contralateral knees were used as controls. Histomorphometric measures of trabecular bone and marrow adipose tissue (MAT) areas were conducted in the epiphysis of the proximal tibia. Knee immobilization for 4, 8, and 16 weeks significantly reduced trabecular bone area by -0.125, -0.139, and -0.161 mm2/mm2, respectively, with corresponding 95 % CIs of [-0.012, -0.239], [-0.006, -0.273], and [-0.101, -0.221]. MAT area significantly increased at 2 and 16 weeks by +0.008 and +0.027 mm2/mm2, respectively, with 95 % CIs of [0.014, 0.002] and [0.039, 0.016]. Remobilization for 8 weeks restored trabecular bone area compared to the contralateral knee and the magnitude of change was significantly greater for 8 and 16 weeks of immobilization with effect sizes of 1.69 and 1.86, respectively. The difference in MAT area between immobilized and contralateral knees were eliminated with remobilization. These results characterize the temporal response of trabecular bone and MAT in the epiphysis of the proximal tibia to joint immobilization and remobilization.
Collapse
|
31
|
Zhou H, Trudel G, Alexeev K, Thomas J, Laneuville O. Hyperplasia and accelerated hypertrophy of marrow adipocytes with knee immobilization were sustained despite remobilization. J Appl Physiol (1985) 2020; 129:701-708. [PMID: 32853104 DOI: 10.1152/japplphysiol.00539.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Skeletal disuse can cause an accumulation of bone marrow adipose tissue (MAT) characterized by a combination of marrow adipocyte hyperplasia and/or hypertrophy. The malleability of MAT accumulation and of the hyperplasia and hypertrophy upon remobilization is unknown. In this study, we showed extensive hyperplasia and accelerated hypertrophy of bone marrow adipocytes in the proximal tibia epiphysis of rat knees immobilized for durations between 1 and 32 wk. Similar histomorphometric measures of adipocytes carried out in unoperated controls allowed distinguishing the effects of immobilization from the effects of aging. Although both knee immobilization and aging led to adipocyte hypertrophy, adipocyte hyperplasia was the hallmark signature effect of immobilization on MAT. Both bone marrow adipocyte hyperplasia and hypertrophy were sustained despite knee remobilization for durations up to four times the duration of immobilization. These results suggest that adipocyte hyperplasia is the predominant mechanism explaining MAT accumulation in skeletal disuse. In this model, the changes were unremitting for the investigated time points. Investigating the cellular and molecular mechanisms of marrow adipocyte mechanoregulation will be important to better understand how adipocytes adapt to changes in mechanical environments.NEW & NOTEWORTHY This longitudinal study elucidates the response of marrow adipose tissue adipocytes in weight-bearing joints to changes in different mechanical environments, and we provide insight on the malleability of the changes over time. In a rat animal model, knee immobilization induced hyperplasia and accelerated the age-dependent hypertrophy of adipocytes. Changes in adipocyte number and size were sustained despite unassisted remobilization. Multimodal distributions of cell size were characteristic of bone marrow adipocytes.
Collapse
Affiliation(s)
- Haodong Zhou
- Department of Biology, Faculty of Science, University of Ottawa, Ontario, Canada.,Bone and Joint Research Laboratory, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Guy Trudel
- Bone and Joint Research Laboratory, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Physical Medicine and Rehabilitation, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada
| | - Konstantin Alexeev
- Department of Biology, Faculty of Science, University of Ottawa, Ontario, Canada.,Bone and Joint Research Laboratory, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Justin Thomas
- Department of Biology, Faculty of Science, University of Ottawa, Ontario, Canada.,Bone and Joint Research Laboratory, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Odette Laneuville
- Department of Biology, Faculty of Science, University of Ottawa, Ontario, Canada.,Bone and Joint Research Laboratory, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
32
|
Wang J, Cui C, Chim YN, Yao H, Shi L, Xu J, Wang J, Wong RMY, Leung KS, Chow SKH, Cheung WH. Vibration and β-hydroxy-β-methylbutyrate treatment suppresses intramuscular fat infiltration and adipogenic differentiation in sarcopenic mice. J Cachexia Sarcopenia Muscle 2020; 11:564-577. [PMID: 31994349 PMCID: PMC7113529 DOI: 10.1002/jcsm.12535] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/26/2019] [Accepted: 12/02/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sarcopenia is an aging-induced deterioration of skeletal muscle mass and function. Low-magnitude high-frequency vibration (LMHFV) was shown to improve muscle functions and β-hydroxy-β-methylbutyrate (HMB) to increase muscle mass and strength. Muscle-derived stem cells (MDSCs) are progenitor cells important for muscle regeneration. We hypothesized that LMHFV and HMB could retard sarcopenia by reducing fat infiltration through inhibiting adipogenesis in MDSCs. METHODS Senescence-accelerated mouse P8 male mice were randomized into control (CTL), HMB, LMHFV (VIB), and combined (COM) groups. Interventions started at age of month 7 and assessed at 1, 2, and 3 months post-intervention by densitometry, histology, and functional tests. In vitro, MDSCs isolated from gastrocnemius of senescence-accelerated mouse P8 mice were characterized, randomized into CTL, VIB, HMB, and COM groups, and assessed by oil red O staining, mRNA, and protein expression. RESULTS At 2 months post-intervention, percentage lean mass of HMB, VIB, and COM groups were significantly higher than CTL group. Twitch, tetanic, and specific tetanic forces of COM group were higher, while specific twitch force of both VIB and COM groups were higher. Grip strength of HMB, VIB, and COM groups were higher. Histologically, both VIB and COM groups presented lower oil red O area than CTL group. Type I muscle fibre in CTL group was higher than HMB, VIB, and COM groups. MDSC were detected in situ by immunofluorescence stain with stem cell antigen-1 signals confirmed with higher β-catenin expression in the COM group. The observations were also confirmed in vitro, MDSCs in the HMB, VIB, and COM groups presented lower adipogenesis vs. the CTL group. β-Catenin mRNA and protein expressions were lower in the CTL group while their relationship was further validated through β-catenin knock-down approach. CONCLUSIONS Our results showed that combined LMHFV and HMB interventions enhanced muscle strength and decreased percentage fat mass and intramuscular fat infiltration as compared with either treatment alone. Additive effect of LMHFV and HMB was demonstrated in β-catenin expression than either treatment in MDSCs and altered cell fate from adipogenesis to myogenesis, leading to inhibition of intramuscular lipid accumulation. Wnt/β-catenin signalling pathway was found to be the predominant regulatory mechanism through which LMHFV and HMB combined treatment suppressed MDSCs adipogenesis.
Collapse
Affiliation(s)
- Jinyu Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Can Cui
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Yu Ning Chim
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Liu Shi
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Jiali Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Ronald Man Yeung Wong
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Kwok-Sui Leung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China
| | - Simon Kwoon-Ho Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, The People's Republic of China
| | - Wing Hoi Cheung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, The People's Republic of China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, The People's Republic of China
| |
Collapse
|
33
|
Putative Receptors for Gravity Sensing in Mammalian Cells: The Effects of Microgravity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10062028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gravity is a constitutive force that influences life on Earth. It is sensed and translated into biochemical stimuli through the so called “mechanosensors”, proteins able to change their molecular conformation in order to amplify external cues causing several intracellular responses. Mechanosensors are widely represented in the human body with important structures such as otholiths in hair cells of vestibular system and statoliths in plants. Moreover, they are also present in the bone, where mechanical cues can cause bone resorption or formation and in muscle in which mechanical stimuli can increase the sensibility for mechanical stretch. In this review, we discuss the role of mechanosensors in two different conditions: normogravity and microgravity, emphasizing their emerging role in microgravity. Microgravity is a singular condition in which many molecular changes occur, strictly connected with the modified gravity force and free fall of bodies. Here, we first summarize the most important mechanosensors involved in normogravity and microgravity. Subsequently, we propose muscle LIM protein (MLP) and sirtuins as new actors in mechanosensing and signaling transduction under microgravity.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The goal of this review is to highlight the deficits in muscle and bone in children with cerebral palsy (CP), discuss the muscle-bone relationship in the CP population, and identify muscle-based intervention strategies that may stimulate an improvement in their bone development. RECENT FINDINGS The latest research suggests that muscle and bone are both severely underdeveloped and weak in children with CP, even in ambulatory children with mild forms of the disorder. The small and low-performing muscles and limited participation in physical activity are likely the major contributors to the poor bone development in children with CP. However, the muscle-bone relationship may be complicated by other factors, such as a high degree of fat and collagen infiltration of muscle, atypical muscle activation, and muscle spasticity. Muscle-based interventions, such as resistance training, vibration, and nutritional supplementation, have the potential to improve bone development in children with CP, especially if they are initiated before puberty. Studies are needed to identify the muscle-related factors with the greatest influence on bone development in children with CP. Identifying treatment strategies that capitalize on the relationship between muscle and bone, while also improving balance, coordination, and physical activity participation, is an important step toward increasing bone strength and minimizing fractures in children with CP.
Collapse
Affiliation(s)
- Christopher M Modlesky
- Department of Kinesiology, University of Georgia, 330 River Road, Room 353, Athens, GA, 30602, USA.
| | - Chuan Zhang
- Department of Kinesiology, University of Georgia, 330 River Road, Room 353, Athens, GA, 30602, USA
| |
Collapse
|
35
|
Horner CB, Maldonado M, Tai Y, Rony RMIK, Nam J. Spatially Regulated Multiphenotypic Differentiation of Stem Cells in 3D via Engineered Mechanical Gradient. ACS APPLIED MATERIALS & INTERFACES 2019; 11:45479-45488. [PMID: 31714732 DOI: 10.1021/acsami.9b17266] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Within the osteochondral interface, cellular and extracellular matrix gradients provide a biomechanical and biochemical niche for homeostatic tissue functions. Postnatal joint loading is critical for the development of such tissue gradients, leading to the formation of functional osteochondral tissues composed of superficial, middle, and deep zones of cartilage, and underlying subchondral bone, in a depth-dependent manner. In this regard, a novel, variable core-shell electrospinning strategy was employed to generate spatially controlled strain gradients within three-dimensional scaffolds under dynamic compressive loading, enabling the local strain-magnitude dependent, multiphenotypic stem cell differentiation. Human mesenchymal stem cells (hMSCs) were cultured in electrospun scaffolds with a linear or biphasic mechanical gradient, which was computationally engineered and experimentally validated. The cell/scaffold constructs were subjected to various magnitudes of dynamic compressive strains in a scaffold depth-dependent manner at a frequency of 1 Hz for 2 h daily for up to 42 days in osteogenic media. Spatially upregulated gene expression of chondrogenic markers (ACAN, COL2A1, PRG4) and glycosaminoglycan deposition was observed in the areas of greater compressive strains. In contrast, osteogenic markers (COL1A1, SPARC, RUNX2) and calcium deposition were downregulated in response to high local compressive strains. Dynamic mechanical analysis showed the maintenance of the engineered mechanical gradients only under dynamic culture conditions, confirming the potent role of biomechanical gradients in developing and maintaining a tissue gradient. These results demonstrate that multiphenotypic differentiation of hMSCs can be controlled by regulating local mechanical microenvironments, providing a novel strategy to recapitulate the gradient structure in osteochondral tissues for successful regeneration of damaged joints in vivo and facile development of interfacial tissue models in vitro.
Collapse
Affiliation(s)
- Christopher B Horner
- Department of Bioengineering , University of California , Riverside , California 92521 , United States
| | - Maricela Maldonado
- Department of Bioengineering , University of California , Riverside , California 92521 , United States
| | - Youyi Tai
- Department of Bioengineering , University of California , Riverside , California 92521 , United States
| | - R M Imtiaz Karim Rony
- Department of Bioengineering , University of California , Riverside , California 92521 , United States
| | - Jin Nam
- Department of Bioengineering , University of California , Riverside , California 92521 , United States
| |
Collapse
|
36
|
Therapeutic effects of whole-body vibration on fracture healing in ovariectomized rats: a systematic review and meta-analysis. Menopause 2019; 26:677-686. [PMID: 30562321 DOI: 10.1097/gme.0000000000001285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Whole-body vibration (WBV), providing cyclic mechanical stimulation, has been used to accelerate fracture healing in preclinical studies. This study aimed to summarize and evaluate the effects of WBV on bone healing in ovariectomized rat models and then analyze its potential effects on fractures in human postmenopausal osteoporosis. METHODS PubMed, EMBASE, Web of Science, China National Knowledge Infrastructure, VIP, SinoMed, and WanFang databases were searched from their inception date to September 2017, and an updated search was conducted in January 2018. Studies that evaluated the effects of WBV on bone healing compared with control groups in ovariectomized rats were included. Two authors selected studies, extracted data, and assessed the methodological quality. Meta-analyses were performed when the same outcomes were reported in two or more studies. RESULTS Nine eligible studies were selected. In treatment groups, callus areas were significantly improved in the first 3 weeks, normalized total bone volume and total tissue volume values increased dramatically at 8 weeks, and the mechanical tests showed a significant difference at the end point of the study. CONCLUSIONS This study suggested that WBV could accelerate callus formation in the early phase of bone healing, promote callus mineralization and maturity in the later phase, and restore mechanical properties of bones.
Collapse
|
37
|
Bourzac C, Bensidhoum M, Pallu S, Portier H. Use of adult mesenchymal stromal cells in tissue repair: impact of physical exercise. Am J Physiol Cell Physiol 2019; 317:C642-C654. [PMID: 31241985 PMCID: PMC6850997 DOI: 10.1152/ajpcell.00530.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Physical exercise (PE) has unquestionable beneficial effects on health, which likely extend into several organ-to-cell physiological processes. At the cell scale, endogenous mesenchymal stromal cells (MSCs) contribute to tissue repair, although their repair capacities may be insufficient in paucicellular or severely damaged tissues. For this reason, MSC transplantation holds great promise for tissue repair. With the goals of understanding if PE has beneficial effects on MSC biology and if PE potentiates their role in tissue repair, we reviewed literature reports regarding the effects of PE on MSC properties (specifically, proliferation, differentiation, and homing) and of a combination of PE and MSC transplantation on tissue repair (specifically neural, cartilage, and muscular tissues). Contradictory results have been reported; interpretation is complicated because various and different species, cell sources, and experimental protocols, specifically exercise programs, have been used. On the basis of these data, the effects of exercise on MSC proliferation and differentiation depend on exercise characteristics (type, intensity, duration, etc.) and on the characteristics of the tissue from which the MSCs were collected. For the in vitro studies, the level of strain (and other details of the mechanical stimulus), the time elapsed between the end of exposure to strain and MSC collection, the age of the donors, as well as the passage number at which the MSCs are evaluated also play a role. The combination of PE and MSC engraftment improves neural, cartilage, and muscular tissue recovery, but it is not clear whether the effects of MSCs and exercise are additive or synergistic.
Collapse
Affiliation(s)
- Celine Bourzac
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Ecole Nationale Vétérinaire d'Alfort, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Maisons-Alfort, France
| | - Morad Bensidhoum
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
| | - Stephane Pallu
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| | - Hugues Portier
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| |
Collapse
|
38
|
Borgiani E, Figge C, Kruck B, Willie BM, Duda GN, Checa S. Age-Related Changes in the Mechanical Regulation of Bone Healing Are Explained by Altered Cellular Mechanoresponse. J Bone Miner Res 2019; 34:1923-1937. [PMID: 31121071 DOI: 10.1002/jbmr.3801] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 04/03/2019] [Accepted: 05/18/2019] [Indexed: 02/05/2023]
Abstract
Increasing age is associated with a reduced bone regeneration potential and increased risk of morbidities and mortality. A reduced bone formation response to mechanical loading has been shown with aging, and it remains unknown if the interplay between aging and mechanical stimuli during regeneration is similar to adaptation. We used a combined in vivo/in silico approach to investigate age-related alterations in the mechanical regulation of bone healing and identified the relative impact of altered cellular function on tissue patterns during the regenerative cascade. To modulate the mechanical environment, femoral osteotomies in adult and elderly mice were stabilized using either a rigid or a semirigid external fixator, and the course of healing was evaluated using histomorphometric and micro-CT analyses at 7, 14, and 21 days post-surgery. Computer models were developed to investigate the influence of the local mechanical environment within the callus on tissue formation patterns. The models aimed to identify the key processes at the cellular level that alter the mechanical regulation of healing with aging. Fifteen age-related biological alterations were investigated on two levels (adult and elderly) with a design of experiments setup. We show a reduced response to changes in fixation stability with age, which could be explained by reduced cellular mechanoresponse, simulated as alteration of the ranges of mechanical stimuli driving mesenchymal stem cell differentiation. Cellular mechanoresponse has been so far widely ignored as a therapeutic target in aged patients. Our data hint to mechanotherapeutics as a potential treatment to enhance bone healing in the elderly. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Edoardo Borgiani
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Christine Figge
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina Kruck
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Georg N Duda
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
39
|
Campsie P, Childs PG, Robertson SN, Cameron K, Hough J, Salmeron-Sanchez M, Tsimbouri PM, Vichare P, Dalby MJ, Reid S. Design, construction and characterisation of a novel nanovibrational bioreactor and cultureware for osteogenesis. Sci Rep 2019; 9:12944. [PMID: 31506561 PMCID: PMC6736847 DOI: 10.1038/s41598-019-49422-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/23/2019] [Indexed: 11/17/2022] Open
Abstract
In regenerative medicine, techniques which control stem cell lineage commitment are a rapidly expanding field of interest. Recently, nanoscale mechanical stimulation of mesenchymal stem cells (MSCs) has been shown to activate mechanotransduction pathways stimulating osteogenesis in 2D and 3D culture. This has the potential to revolutionise bone graft procedures by creating cellular graft material from autologous or allogeneic sources of MSCs without using chemical induction. With the increased interest in mechanical stimulation of cells and huge potential for clinical use, it is apparent that researchers and clinicians require a scalable bioreactor system that provides consistently reproducible results with a simple turnkey approach. A novel bioreactor system is presented that consists of: a bioreactor vibration plate, calibrated and optimised for nanometre vibrations at 1 kHz, a power supply unit, which supplies a 1 kHz sine wave signal necessary to generate approximately 30 nm of vibration amplitude, and custom 6-well cultureware with toroidal shaped magnets incorporated in the base of each well for conformal attachment to the bioreactor’s magnetic vibration plate. The cultureware and vibration plate were designed using finite element analysis to determine the modal and harmonic responses, and validated by interferometric measurement. This helps ensure that the vibration plate and cultureware, and thus collagen and MSCs, all move as a rigid body, avoiding large deformations close to the resonant frequency of the vibration plate and vibration damping beyond the resonance. Assessment of osteogenic protein expression was performed to confirm differentiation of MSCs after initial biological experiments with the system, as well as atomic force microscopy of the 3D gel constructs during vibrational stimulation to verify that strain hardening of the gel did not occur. This shows that cell differentiation was the result of the nanovibrational stimulation provided by the bioreactor alone, and that other cell differentiating factors, such as stiffening of the collagen gel, did not contribute.
Collapse
Affiliation(s)
- Paul Campsie
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Peter G Childs
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Shaun N Robertson
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Kenny Cameron
- School of Computing, Engineering and Physical Sciences, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - James Hough
- SUPA Institute for Gravitational Research, School of Physics and Astronomy, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Penelope M Tsimbouri
- Centre for the Cellular Microenvironment, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Parag Vichare
- School of Computing, Engineering and Physical Sciences, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| | - Stuart Reid
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK.
| |
Collapse
|
40
|
Applying vibration in early postmenopausal osteoporosis promotes osteogenic differentiation of bone marrow-derived mesenchymal stem cells and suppresses postmenopausal osteoporosis progression. Biosci Rep 2019; 39:BSR20191011. [PMID: 31406012 PMCID: PMC6722487 DOI: 10.1042/bsr20191011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/29/2019] [Accepted: 08/08/2019] [Indexed: 02/05/2023] Open
Abstract
We aimed to evaluate whether applying low magnitude vibration (LMV) in early postmenopausal osteoporosis (PMO) suppresses its progression, and to investigate underlying mechanisms. Rats were randomly divided into Sham (Sham-operated), Sham+V, OVX (ovariectomized), OVX+E2 (estradiol benzoate), OVX+V (LMV at 12–20 weeks postoperatively), and OVX+Vi (LMV at 1–20 weeks postoperatively) groups. LMV was applied for 20 min once daily for 5 days weekly. V rats were loaded with LMV at 12–20 weeks postoperatively. Vi rats were loaded with LMV at 1–20 weeks postoperatively. Estradiol (E2) rats were intramuscularly injected at 12–20 weeks postoperatively once daily for 3 days. The bone mineral densities (BMDs), biomechanical properties, and histomorphological parameters of tibiae were analyzed. In vitro, rat bone marrow-derived mesenchymal stem cells (rBMSCs) were subjected to LMV for 30 min daily for 5 days, or 17β-E2 with or without 1-day pretreatment of estrogen receptor (ER) inhibitor ICI 182,780 (ICI). The mRNA and protein expresion were performed. Data showed that LMV increased BMD, bone strength, and bone mass of rats, and the effects of Vi were stronger than those of E2. In vitro, LMV up-regulated the mRNA and protein expressions of Runx2, Osx, Col I, and OCN and down-regulated PPARγ, compared with E2. The effects of both LMV and E2 on rBMSCs were inhibited by ICI. Altogether, LMV in early PMO suppresses its progression, which is associated with osteogenic differentiation of rBMSCs via up-regulation of ERα and activation of the canonical Wnt pathway. LMV may therefore be superior to E2 for the suppression of PMO progression.
Collapse
|
41
|
Bizelli-Silveira C, Abildtrup LA, Spin-Neto R, Foss M, Søballe K, Kraft DCE. Strontium enhances proliferation and osteogenic behavior of bone marrow stromal cells of mesenchymal and ectomesenchymal origins in vitro. Clin Exp Dent Res 2019; 5:541-550. [PMID: 31687189 PMCID: PMC6820574 DOI: 10.1002/cre2.221] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Obejective To investigate the effect of increasing Strontium (Sr) concentrations on the growth and osteogenic behavior of human bone marrow stromal cells (BMSCs) from mesenchymal (i.e., fibula) and ectomesenchymal (i.e., mandible) embryonic origins. Materials and methods Fibula and mandible BMSCs were cultured in media without (Ctrl) or with Sr in four diverse concentrations: Sr1, 11.3 × 10−3 mg/L, human seric physiological level; Sr2, 13 mg/L, human seric level after strontium ranelate treatment; Sr3, 130 mg/L, and Sr4, 360 mg/L. Proliferation rate (1, 3, and 7 days), osteogenic behavior (alkaline phosphatase [ALP] activity, 7 and 14 days; expression of osteogenic genes (ALP, osteopontin, and osteocalcin at 7, 14, and 21 days), and formation of mineralized nodules (14 and 21 days) of the BMSCs were assessed. Data was compared group‐ and period‐wise using analysis of variance tests. Results Fibula and mandible BMSCs cultured with Sr4 showed increased proliferation rate, and osteocalcin and osteopontin gene expression together with more evident formation of mineralized nodules, compared all other Sr concentrations. For both cell populations, Sr4 led to lower ALP activity, and ALP gene expression, compared with the other Sr concentrations. Conclusion BMSCs from mesenchymal (i.e., fibula) and ectomesenchymal (i.e., mandible) embryonic origins showed increased cellular proliferation and osteogenic behavior when cultured with Sr4, in vitro.
Collapse
Affiliation(s)
- Carolina Bizelli-Silveira
- Department of Dentistry and Oral Health, Faculty of Health Aarhus University Aarhus Denmark.,Department of Orthopaedic Surgery Aarhus University Hospital Aarhus Denmark
| | - Lisbeth Ann Abildtrup
- Department of Dentistry and Oral Health, Faculty of Health Aarhus University Aarhus Denmark
| | - Rubens Spin-Neto
- Department of Dentistry and Oral Health, Faculty of Health Aarhus University Aarhus Denmark
| | - Morten Foss
- Interdisciplinary Nanoscience Center (iNANO), Department of Physics and Astronomy, Faculty of Science and Technology Aarhus University Aarhus Denmark
| | - Kjeld Søballe
- Department of Orthopaedic Surgery Aarhus University Hospital Aarhus Denmark
| | | |
Collapse
|
42
|
Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK, Guise TA, Rubin J, Rubin CT. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol 2019; 15:339-355. [PMID: 30814687 PMCID: PMC6520125 DOI: 10.1038/s41574-019-0170-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoporosis, a condition of skeletal decline that undermines quality of life, is treated with pharmacological interventions that are associated with poor adherence and adverse effects. Complicating efforts to improve clinical outcomes, the incidence of obesity is increasing, predisposing the population to a range of musculoskeletal complications and metabolic disorders. Pharmacological management of obesity has yet to deliver notable reductions in weight and debilitating complications are rarely avoided. By contrast, exercise shows promise as a non-invasive and non-pharmacological method of regulating both osteoporosis and obesity. The principal components of exercise - mechanical signals - promote bone and muscle anabolism while limiting formation and expansion of fat mass. Mechanical regulation of bone and marrow fat might be achieved by regulating functions of differentiated cells in the skeletal tissue while biasing lineage selection of their common progenitors - mesenchymal stem cells. An inverse relationship between adipocyte versus osteoblast fate selection from stem cells is implicated in clinical conditions such as childhood obesity and increased marrow adiposity in type 2 diabetes mellitus, as well as contributing to skeletal frailty. Understanding how exercise-induced mechanical signals can be used to improve bone quality while decreasing fat mass and metabolic dysfunction should lead to new strategies to treat chronic diseases such as osteoporosis and obesity.
Collapse
Affiliation(s)
- Gabriel M Pagnotti
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- College of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Laura E Wright
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Theresa A Guise
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
43
|
Touchstone H, Bryd R, Loisate S, Thompson M, Kim S, Puranam K, Senthilnathan AN, Pu X, Beard R, Rubin J, Alwood J, Oxford JT, Uzer G. Recovery of stem cell proliferation by low intensity vibration under simulated microgravity requires LINC complex. NPJ Microgravity 2019; 5:11. [PMID: 31123701 PMCID: PMC6520402 DOI: 10.1038/s41526-019-0072-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSC) rely on their ability to integrate physical and spatial signals at load bearing sites to replace and renew musculoskeletal tissues. Designed to mimic unloading experienced during spaceflight, preclinical unloading and simulated microgravity models show that alteration of gravitational loading limits proliferative activity of stem cells. Emerging evidence indicates that this loss of proliferation may be linked to loss of cellular cytoskeleton and contractility. Low intensity vibration (LIV) is an exercise mimetic that promotes proliferation and differentiation of MSCs by enhancing cell structure. Here, we asked whether application of LIV could restore the reduced proliferative capacity seen in MSCs that are subjected to simulated microgravity. We found that simulated microgravity (sMG) decreased cell proliferation and simultaneously compromised cell structure. These changes included increased nuclear height, disorganized apical F-actin structure, reduced expression, and protein levels of nuclear lamina elements LaminA/C LaminB1 as well as linker of nucleoskeleton and cytoskeleton (LINC) complex elements Sun-2 and Nesprin-2. Application of LIV restored cell proliferation and nuclear proteins LaminA/C and Sun-2. An intact LINC function was required for LIV effect; disabling LINC functionality via co-depletion of Sun-1, and Sun-2 prevented rescue of cell proliferation by LIV. Our findings show that sMG alters nuclear structure and leads to decreased cell proliferation, but does not diminish LINC complex mediated mechanosensitivity, suggesting LIV as a potential candidate to combat sMG-induced proliferation loss.
Collapse
Affiliation(s)
- H. Touchstone
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID 83725 USA
| | - R. Bryd
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID 83725 USA
| | - S. Loisate
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID 83725 USA
| | - M. Thompson
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID 83725 USA
| | - S. Kim
- Department of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599 USA
| | - K. Puranam
- Department of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599 USA
| | - A. N. Senthilnathan
- Department of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599 USA
| | - X. Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | - R. Beard
- Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | - J. Rubin
- Department of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599 USA
| | - J. Alwood
- Space Biosciences Division, NASA-Ames Research Center, Mountain View, CA 94035 USA
| | - J. T. Oxford
- Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | - G. Uzer
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID 83725 USA
| |
Collapse
|
44
|
Abstract
For mechanical force to induce changes in cellular behaviors, two main processes are inevitable; perception of the force and response to it. Perception of mechanical force by cells, or mechanosensing, requires mechanical force-induced conformational changes in mechanosensors. For this, at least one end of the mechanosensors should be anchored to relatively fixed structures, such as extracellular matrices or the cytoskeletons, while the other end should be pulled along the direction of the mechanical force. Alternatively, mechanosensors may be positioned in lipid bilayers, so that conformational changes in the embedded sensors can be induced by mechanical force-driven tension in the lipid bilayer. Responses to mechanical force by cells, or mechanotransduction, require translation of such mechanical force-induced conformational changes into biochemical signaling. For this, protein-protein interactions or enzymatic activities of mechanosensors should be modulated in response to force-induced structural changes. In the last decade, several molecules that met the required criteria of mechanosensors have been identified and proven to directly sense mechanical force. The present review introduces examples of such mechanosensors and summarizes their mechanisms of action.
Collapse
Affiliation(s)
- Chul-Gyun Lim
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Jiyoung Jang
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Chungho Kim
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
45
|
Kim J, Li B, Scheideler OJ, Kim Y, Sohn LL. Visco-Node-Pore Sensing: A Microfluidic Rheology Platform to Characterize Viscoelastic Properties of Epithelial Cells. iScience 2019; 13:214-228. [PMID: 30870780 PMCID: PMC6416673 DOI: 10.1016/j.isci.2019.02.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/26/2019] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Viscoelastic properties of cells provide valuable information regarding biological or clinically relevant cellular characteristics. Here, we introduce a new, electronic-based, microfluidic platform-visco-node-pore sensing (visco-NPS)-which quantifies cellular viscoelastic properties under periodic deformation. We measure the storage (G') and loss (G″) moduli (i.e., elasticity and viscosity, respectively) of cells. By applying a wide range of deformation frequencies, our platform quantifies the frequency dependence of viscoelastic properties. G' and G″ measurements show that the viscoelastic properties of malignant breast epithelial cells (MCF-7) are distinctly different from those of non-malignant breast epithelial cells (MCF-10A). With its sensitivity, visco-NPS can dissect the individual contributions of different cytoskeletal components to whole-cell mechanical properties. Moreover, visco-NPS can quantify the mechanical transitions of cells as they traverse the cell cycle or are initiated into an epithelial-mesenchymal transition. Visco-NPS identifies viscoelastic characteristics of cell populations, providing a biophysical understanding of cellular behavior and a potential for clinical applications.
Collapse
Affiliation(s)
- Junghyun Kim
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA, USA
| | - Brian Li
- Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA
| | - Olivia J Scheideler
- Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA
| | - Youngbin Kim
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA, USA
| | - Lydia L Sohn
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA, USA; Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA.
| |
Collapse
|
46
|
Lee IC, Wu HJ, Liu HL. Dual-Frequency Ultrasound Induces Neural Stem/Progenitor Cell Differentiation and Growth Factor Utilization by Enhancing Stable Cavitation. ACS Chem Neurosci 2019; 10:1452-1461. [PMID: 30608667 DOI: 10.1021/acschemneuro.8b00483] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) have the potential to serve as the basic materials for treating severe neural diseases and injuries. Ultrasound exposure is an effective therapy for nonunion fractures and healing fresh wounds through an easy and noninvasive application. According to the results of our preliminary study, low-intensity ultrasound (LIUS) promotes the attachment and differentiation of NSPCs. However, the parameters of and mechanisms by which LIUS induces NSPC differentiation remain unclear. To the best of our knowledge, no published studies have reported and compared the biological effects of dual-frequency and single-frequency LIUS on NSPCs. The purpose of this study is to systematically compare several LIUS parameters, including single-frequency, single-transducer dual-frequency ultrasound, burst, and continuous cycling stimulation at several intensities. Furthermore, synergistic effects of single-/dual-frequency LIUS combined with neural growth factor addition on NSPCs were also evaluated. Based on the results of the cytotoxicity assay, low-intensity (40 kPa) ultrasound does not damage NSPCs compared with that observed in the control group. The morphology and immunostaining results show that all experimental groups exposed to ultrasound exhibit neurite outgrowth and NSPC differentiation. In particular, dual-frequency ultrasound promotes NSPCs differentiation to a greater extent than single-frequency ultrasound. In addition, more complicated and denser neural networks are observed in the dual-frequency group. Neural growth factor addition increased the percentage of neurons formed, particularly in the groups stimulated with ultrasound. Among these groups, the dual-frequency group exhibited significant differences in the percentage of differentiated neurons compared with the single-frequency group. This study may the first to prove that dual-frequency LIUS exposure further enhances NSPC differentiation and the utilization of growth factors than single-frequency LIUS. Moreover, the result also revealed that dual-frequency ultrasound generated higher calcium ion influx and extended the channel opening time. A potential explanation is that dual-frequency ultrasound generates more stable cavitation than single-frequency LIUS, which may stimulate cell membrane mechanochannels and enhance calcium ion influx but does not damage them. This in vitro study may serve as a useful alternative for ultrasound therapy.
Collapse
Affiliation(s)
- I-Chi Lee
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
- Neurosurgery Department, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hui-Ju Wu
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
47
|
Hawkes CP, Mostoufi-Moab S. Fat-bone interaction within the bone marrow milieu: Impact on hematopoiesis and systemic energy metabolism. Bone 2019; 119:57-64. [PMID: 29550266 PMCID: PMC6139083 DOI: 10.1016/j.bone.2018.03.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/13/2018] [Indexed: 12/23/2022]
Abstract
The relationship between fat, bone and systemic metabolism is a growing area of scientific interest. Marrow adipose tissue is a well-recognized component of the bone marrow milieu and is metabolically distinct from current established subtypes of adipose tissue. Despite recent advances, the functional significance of marrow adipose tissue is still not clearly delineated. Bone and fat cells share a common mesenchymal stem cell (MSC) within the bone marrow, and hormones and transcription factors such as growth hormone, leptin, and peroxisomal proliferator-activated receptor γ influence MSC differentiation into osteoblasts or adipocytes. MSC osteogenic potential is more vulnerable than adipogenic potential to radiation and chemotherapy, and this confers a risk for an abnormal fat-bone axis in survivors following cancer therapy and bone marrow transplantation. This review provides a summary of data from animal and human studies describing the relationship between marrow adipose tissue and hematopoiesis, bone mineral density, bone strength, and metabolic function. The significance of marrow adiposity in other metabolic disorders such as osteoporosis, diabetes mellitus, and estrogen and growth hormone deficiency are also discussed. We conclude that marrow adipose tissue is an active endocrine organ with important metabolic functions contributing to bone energy maintenance, osteogenesis, bone remodeling, and hematopoiesis. Future studies on the metabolic role of marrow adipose tissue may provide the critical insight necessary for selecting targeted therapeutic interventions to improve altered hematopoiesis and augment skeletal remodeling in cancer survivors.
Collapse
Affiliation(s)
- C P Hawkes
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - S Mostoufi-Moab
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, USA; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, USA; Perelman School of Medicine, Department of Pediatrics, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
48
|
Kang X, Sun Y, Zhang Z. Identification of key transcription factors - gene regulatory network related with osteogenic differentiation of human mesenchymal stem cells based on transcription factor prognosis system. Exp Ther Med 2019; 17:2113-2122. [PMID: 30783479 PMCID: PMC6364222 DOI: 10.3892/etm.2019.7170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) have the capacity to differentiate into fabricate cartilage, muscle, marrow stroma, tendon/ligament, fat, and other connective tissues, providing a potential source for tissue regeneration. The aim of this study was to find the key transcription factors (TFs), which regulated osteogenic differentiation of hMSCs. In this study, three methods were performed to find the key TFs, which included enrichment analysis, direct impact value and indirect impact value. We used the patient and public involvements (PPI) network to integrate the results of the above methods for analysis. Then, we compared the osteoblast data to the control group on days 1, 3 and 7. Finally, we found the combination of the optimal and vital 30 TFs related to osteogenic differentiation. TFs FOS, SOX9 and EP300 were commonly expressed in 3 different days in the osteogenic lineages and presented in the PPI network at relatively high degrees. Moreover, TFs CREBBP, ESR1 and EGR1 also presented high effects on the 1st, 3rd and 7th day. The constructed network gives us a more comprehensive understanding of the mechanism of osteogenesis of hMSCs.
Collapse
Affiliation(s)
- Xuefeng Kang
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150030, P.R. China
| | - Yong Sun
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150030, P.R. China
| | - Zhao Zhang
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150030, P.R. China
| |
Collapse
|
49
|
Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9010174] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies.
Collapse
|
50
|
Lin J, Anopas D, Milbreta U, Lin PH, Chin JS, Zhang N, Wee SK, Tow A, Ang WT, Chew SY. Regenerative rehabilitation: exploring the synergistic effects of rehabilitation and implantation of a bio-functional scaffold in enhancing nerve regeneration. Biomater Sci 2019; 7:5150-5160. [DOI: 10.1039/c9bm01095e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Combinatorial approach of rehabilitation and regeneration is essential for functional recovery.
Collapse
Affiliation(s)
- Junquan Lin
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| | - Dollaporn Anopas
- School of Mechanical and Aerospace Engineering
- Nanyang Technological University
- Singapore
| | - Ulla Milbreta
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| | - Po Hen Lin
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
- NTU Institute for Health Technologies (Health Tech NTU)
- Interdisciplinary Graduate School
| | - Na Zhang
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
| | - Seng Kwee Wee
- Department of Rehabilitation Medicine
- Tan Tock Seng Hospital
- Singapore
| | - Adela Tow
- Department of Rehabilitation Medicine
- Tan Tock Seng Hospital
- Singapore
| | - Wei Tech Ang
- School of Mechanical and Aerospace Engineering
- Nanyang Technological University
- Singapore
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore
- Lee Kong Chian School of Medicine
- Nanyang Technological University
| |
Collapse
|