1
|
Shin YK, Heo JH, Lee JY, Park YJ, Cho SR. Collagen-binding peptide reverses bone loss in a mouse model of cerebral palsy based on clinical databases. Ann Phys Rehabil Med 2020; 64:101445. [PMID: 33130040 DOI: 10.1016/j.rehab.2020.09.009] [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: 02/17/2020] [Revised: 08/06/2020] [Accepted: 09/27/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Individuals with cerebral palsy (CP) experience bone loss due to impaired weight bearing. Despite serious complications, there is no standard medication. OBJECTIVE To develop a new pharmacological agent, we performed a series of studies. The primary aim was to develop an animal model of CP to use our target medication based on transcriptome analysis of individuals with CP. The secondary aim was to show the therapeutic capability of collagen-binding peptide (CBP) in reversing bone loss in the CP mouse model. METHODS A total of 119 people with CP and 13 healthy adults participated in the study and 140 mice were used for the behavioral analysis and discovery of therapeutic effects in the preclinical study. The mouse model of CP was induced by hypoxic-ischemic brain injury. Inclusion and exclusion criteria were established for CBP medication in the CP mouse model with bone loss. RESULTS On the basis of clinical outcomes showing insufficient mechanical loading from non-ambulatory function and that underweight mainly affects bone loss in adults with CP, we developed a mouse model of CP with bone loss. Injury severity and body weight mainly affected bone loss in the CP mouse model. Transcriptome analysis showed SPP1 expression downregulated in adults with CP who showed lower bone density than healthy controls. Therefore, a synthesized CBP was administered to the mouse model. Trabecular thickness, total collagen and bone turnover activity increased with CBP treatment as compared with the saline control. Immunohistochemistry showed increased immunoreactivity of runt-related transcription factor 2 and osteocalcin, so the CBP participated in osteoblast differentiation. CONCLUSIONS This study can provide a scientific basis for a promising translational approach for developing new anabolic CBP medication to treat bone loss in individuals with CP.
Collapse
Affiliation(s)
- Yoon-Kyum Shin
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea
| | - Jeong Hyun Heo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea; Graduate Program of NanoScience and Technology, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea
| | - Jue Yeon Lee
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), 03080 Seoul, Republic of Korea
| | - Yoon-Jeong Park
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), 03080 Seoul, Republic of Korea; Department of Dental Regenerative Biotechnology, School of Dentistry, Seoul National University, 03080 Seoul, Republic of Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea; Graduate Program of NanoScience and Technology, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea; Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, 03722 Seoul, Republic of Korea.
| |
Collapse
|
2
|
He Z, Nie P, Lu J, Ling Y, Guo J, Zhang B, Hu J, Liao J, Gu J, Dai B, Feng Z. Less mechanical loading attenuates osteoarthritis by reducing cartilage degeneration, subchondral bone remodelling, secondary inflammation, and activation of NLRP3 inflammasome. Bone Joint Res 2020; 9:731-741. [PMID: 33399476 PMCID: PMC7640939 DOI: 10.1302/2046-3758.910.bjr-2019-0368.r2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aims Osteoarthritis (OA) is a disabling joint disorder and mechanical loading is an important pathogenesis. This study aims to investigate the benefits of less mechanical loading created by intermittent tail suspension for knee OA. Methods A post-traumatic OA model was established in 20 rats (12 weeks old, male). Ten rats were treated with less mechanical loading through intermittent tail suspension, while another ten rats were treated with normal mechanical loading. Cartilage damage was determined by gross appearance, Safranin O/Fast Green staining, and immunohistochemistry examinations. Subchondral bone changes were analyzed by micro-CT and tartrate-resistant acid phosphatase (TRAP) staining, and serum inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay (ELISA). Results Our radiographs showed that joint space was significantly enlarged in rats with less mechanical loading. Moreover, cartilage destruction was attenuated in the less mechanical loading group with lower histological damage scores, and lower expression of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5, matrix metalloproteinase (MMP)-3, and MMP-13. In addition, subchondral bone abnormal changes were ameliorated in OA rats with less mechanical loading, as reduced bone mineral density (BMD), bone volume/tissue volume (BV/TV), and number of osteophytes and osteoclasts in the subchondral bone were observed. Finally, the level of serum inflammatory cytokines was significantly downregulated in the less mechanical loading group compared with the normal mechanical loading group, as well as the expression of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3), caspase-1, and interleukin 1β (IL-1β) in the cartilage. Conclusion Less mechanical loading alleviates cartilage destruction, subchondral bone changes, and secondary inflammation in OA joints. This study provides fundamental insights into the benefit of non-weight loading rest for patients with OA. Cite this article: Bone Joint Res 2020;9(10):731–741.
Collapse
Affiliation(s)
- Zhennian He
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Pengfei Nie
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Jianli Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yong Ling
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Jian Guo
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Bin Zhang
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Jianghua Hu
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Jiawei Liao
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Jie Gu
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Bo Dai
- Department of Orthopedic Surgery, Beilun District People's Hospital, Ningbo, China
| | - Zhiyun Feng
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Matthews BG, Wee NKY, Widjaja VN, Price JS, Kalajzic I, Windahl SH. αSMA Osteoprogenitor Cells Contribute to the Increase in Osteoblast Numbers in Response to Mechanical Loading. Calcif Tissue Int 2020; 106:208-217. [PMID: 31673746 PMCID: PMC6995756 DOI: 10.1007/s00223-019-00624-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 10/11/2019] [Indexed: 01/11/2023]
Abstract
Bone is a dynamic tissue that site-specifically adapts to the load that it experiences. In response to increasing load, the cortical bone area is increased, mainly through enhanced periosteal bone formation. This increase in area is associated with an increase in the number of bone-forming osteoblasts; however, the origin of the cells involved remains unclear. Alpha-smooth muscle actin (αSMA) is a marker of early osteoprogenitor cells in the periosteum, and we hypothesized that the new osteoblasts that are activated by loading could originate from αSMA-expressing cells. Therefore, we used an in vivo fate-mapping approach in an established axial loading model to investigate the role of αSMA-expressing cells in the load-induced increase in osteoblasts. Histomorphometric analysis was applied to measure the number of cells of different origin on the periosteal surface in the most load-responsive region of the mouse tibia. A single loading session failed to increase the number of periosteal αSMA-expressing cells and osteoblasts. However, in response to multiple episodes of loading, the caudal, but not the cranial, periosteal surface was lined with an increased number of osteoblasts originating from αSMA-expressing cells 5 days after the initial loading session. The proportion of osteoblasts derived from αSMA-labeled progenitors increased by 70% (p < 0.05), and the proportion of αSMA-labeled cells that had differentiated into osteoblasts was doubled. We conclude that αSMA-expressing osteoprogenitors can differentiate and contribute to the increase in periosteal osteoblasts induced by mechanical loading in a site-specific manner.
Collapse
Affiliation(s)
- B G Matthews
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - N K Y Wee
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA
| | - V N Widjaja
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - J S Price
- School of Veterinary Sciences, University of Bristol, Bristol, UK
- Royal Agricultural University, Cirencester, UK
| | - I Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA
| | - S H Windahl
- School of Veterinary Sciences, University of Bristol, Bristol, UK.
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
4
|
Metzger CE, Brezicha JE, Elizondo JP, Narayanan SA, Hogan HA, Bloomfield SA. Differential responses of mechanosensitive osteocyte proteins in fore- and hindlimbs of hindlimb-unloaded rats. Bone 2017; 105:26-34. [PMID: 28782619 DOI: 10.1016/j.bone.2017.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/12/2017] [Accepted: 08/02/2017] [Indexed: 12/28/2022]
Abstract
Osteocytes are believed to be the primary mechanosensors of bone tissue, signaling to osteoblasts and osteoclasts by releasing specific proteins. Sclerostin, interleukin-6 (IL-6), and insulin-like growth factor-I (IGF-I) are osteocyte proteins that signal to osteoblasts. The primary objective of this study was to determine if osteocyte protein response to mechanical unloading is restricted to the unloaded bone using the hindlimb unloading (HU) rodent model. We also examined tumor necrosis factor-α (TNF-α) due to its interactions with all three osteocyte proteins. We hypothesized that unloaded hindlimb cancellous bone would have an altered osteocyte protein (sclerostin, IL-6, and IGF-I) response compared to controls, while the response in the weight-bearing forelimb would not differ from ambulating controls. Male Sprague Dawley rats (7-mo old) experienced either HU (n=7) or normal cage activity (CON; n=7) for 28days. The unloaded distal femur and the weight-bearing proximal humerus were compared in HU vs CON. Metaphyseal bone density was reduced in the HU rats' hindlimb, but not in the proximal humerus, compared to CON values. Osteocyte density was 30% lower in the HU distal femur, but not different from CON in the proximal humerus. %Sclerostin+osteocytes in the distal femur were higher in HU compared to CON, but lower in the proximal humerus. Both %IGF-I+ and %IL-6+ osteocytes were lower in the distal femur for HU, but higher in the proximal humerus for HU. Osterix surface, a marker of osteoblasts, was lower in HU in the distal femur; however, the proximal humerus had more %osterix+surface in HU. In HU %Cathepsin K+ surface, a marker of osteoclasts, was higher in the distal femur and lower in the proximal humerus. %TNF-α+osteocytes were no different from CON in either bone site. HU proximal humerus osteocyte protein responses of sclerostin, IL-6, and IGF-I changed in the opposite direction as observed in the distal femur within the same animal. The opposite response of osteocyte proteins and osteoblast surface in hind- and forelimb bones within the same animal suggests that, while osteocytes in the unloaded hindlimb sense a lack of mechanical strain, osteocytes in the weight-bearing forelimb in HU animals sense some increase in local strain and generate molecular signaling to osteoblasts.
Collapse
Affiliation(s)
- Corinne E Metzger
- Department of Health and Kinesiology, Texas A&M University, College Station, TX 77843, United States
| | - Jessica E Brezicha
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Jon P Elizondo
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, United States
| | - S Anand Narayanan
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX 76504, United States
| | - Harry A Hogan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States; Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Susan A Bloomfield
- Department of Health and Kinesiology, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
5
|
Shao XR, Lin SY, Peng Q, Shi SR, Li XL, Zhang T, Lin YF. Effect of tetrahedral DNA nanostructures on osteogenic differentiation of mesenchymal stem cells via activation of the Wnt/β-catenin signaling pathway. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1809-1819. [PMID: 28259801 DOI: 10.1016/j.nano.2017.02.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/10/2017] [Accepted: 02/15/2017] [Indexed: 02/05/2023]
Abstract
Adipose-derived stem cells (ADSCs) are considered to be ideal stem cell sources for bone regeneration owing to their ability to differentiate into osteo-like cells. Therefore, they have attracted increasing attention in recent years. Tetrahedral DNA nanostructures (TDNs), a new type of DNA-based biomaterials, have shown great potential for biomedical applications. In the present work, we aimed to investigate the role played by TDNs in osteogenic differentiation and proliferation of ADSCs and tried to explore if the canonical Wnt signal pathway could be the vital biological mechanism driving these cellular responses. Upon exposure to TDNs, ADSCs proliferation and osteogenic differentiation were significantly enhanced, accompanied by the up-regulation of genes correlated with the Wnt/β-catenin pathway. In conclusion, our results indicate that TDNs are crucial regulators of the increase in osteogenic potential and ADSCs proliferation, and this noteworthy discovery could provide a promising novel approach toward ADSCs-based bone defect regeneration.
Collapse
Affiliation(s)
- Xiao-Ru Shao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shi-Yu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Si-Rong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao-Long Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yun-Feng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Qin W, Zhao W, Li X, Peng Y, Harlow LM, Li J, Qin Y, Pan J, Wu Y, Ran L, Ke HZ, Cardozo CP, Bauman WA. Mice with sclerostin gene deletion are resistant to the severe sublesional bone loss induced by spinal cord injury. Osteoporos Int 2016; 27:3627-3636. [PMID: 27436301 DOI: 10.1007/s00198-016-3700-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/01/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Bone loss after spinal cord injury (SCI) is rapid, severe, and refractory to interventions studied to date. Mice with sclerostin gene deletion are resistant to the severe sublesional bone loss induced by SCI, further indicating pharmacological inhibition of sclerostin may represent a promising novel approach to this challenging medical problem. INTRODUCTION The bone loss secondary to spinal cord injury (SCI) is associated with several unique pathological features, including the permanent immobilization, neurological dysfunction, and systemic hormonal alternations. It remains unclear how these complex pathophysiological changes are linked to molecular alterations that influence bone metabolism in SCI. Sclerostin is a key negative regulator of bone formation and bone mass. We hypothesized that sclerostin could function as a major mediator of bone loss following SCI. METHODS To test this hypothesis, 10-week-old female sclerostin knockout (SOST KO) and wild type (WT) mice underwent complete spinal cord transection or laminectomy (Sham). RESULTS At 8 weeks after SCI, substantial loss of bone mineral density was observed at the distal femur and proximal tibia in WT mice but not in SOST KO mice. By μCT, trabecular bone volume of the distal femur was markedly decreased by 64 % in WT mice after SCI. In striking contrast, there was no significant reduction of bone volume in SOST KO/SCI mice compared with SOST KO/sham. Histomorphometric analysis of trabecular bone revealed that the significant reduction in bone formation rate following SCI was observed in WT mice but not in SOST KO mice. Moreover, SCI did not alter osteoblastogenesis of marrow stromal cells in SOST KO mice. CONCLUSION Our findings demonstrate that SOST KO mice were protected from the major sublesional bone loss that invariably follows SCI. The evidence indicates that sclerostin is an important mediator of the marked sublesional bone loss after SCI, and that pharmacological inhibition of sclerostin may represent a promising novel approach to this challenging clinical problem.
Collapse
Affiliation(s)
- W Qin
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA.
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - W Zhao
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - X Li
- Amgen Inc, Thousand Oaks, CA, USA
| | - Y Peng
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - L M Harlow
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - J Li
- Indiana University Purdue University, Indianapolis, IN, USA
| | - Y Qin
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - J Pan
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
| | - Y Wu
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute of Gene Engineering Animal Models for Human Diseases, Dalian Medical University, Dalian, China
| | - L Ran
- Institute of Gene Engineering Animal Models for Human Diseases, Dalian Medical University, Dalian, China
| | | | - C P Cardozo
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W A Bauman
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Roa, Bronx, NY, 10468, USA
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
7
|
Li B, Sankaran JS, Judex S. Trabecular and Cortical Bone of Growing C3H Mice Is Highly Responsive to the Removal of Weightbearing. PLoS One 2016; 11:e0156222. [PMID: 27223115 PMCID: PMC4880346 DOI: 10.1371/journal.pone.0156222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/11/2016] [Indexed: 01/08/2023] Open
Abstract
Genetic make-up strongly influences the skeleton’s susceptibility to the loss of weight bearing with some inbred mouse strains experiencing great amounts of bone loss while others lose bone at much smaller rates. At young adulthood, female inbred C3H/HeJ (C3H) mice are largely resistant to catabolic pressure induced by unloading. Here, we tested whether the depressed responsivity to unloading is inherent to the C3H genetic make-up or whether a younger age facilitates a robust skeletal response to unloading. Nine-week-old, skeletally immature, female C3H mice were subjected to 3wk of hindlimb unloading (HLU, n = 12) or served as normal baseline controls (BC, n = 10) or age-matched controls (AC, n = 12). In all mice, cortical and trabecular architecture of the femur, as well as levels of bone formation and resorption, were assessed with μCT, histomorphometry, and histology. Changes in bone marrow progenitor cell populations were determined with flow cytometry. Following 21d of unloading, HLU mice had 52% less trabecular bone in the distal femur than normal age-matched controls. Reflecting a loss of trabecular tissue compared to baseline controls, trabecular bone formation rates (BFR/BS) in HLU mice were 40% lower than in age-matched controls. Surfaces undergoing osteoclastic resorption were not significantly different between groups. In the mid-diaphysis, HLU inhibited cortical bone growth leading to 14% less bone area compared to age-matched controls. Compared to AC, BFR/BS of HLU mice were 53% lower at the endo-cortical surface and 49% lower at the periosteal surface of the mid-diaphysis. The enriched osteoprogenitor cell population (OPC) comprised 2% of the bone marrow stem cells in HLU mice, significantly different from 3% OPC in the AC group. These data show that bone tissue in actively growing C3H mice is lost rapidly, or fails to grow, during the removal of functional weight bearing—in contrast to the insignificant response previously demonstrated in female young adult C3H mice. Thus, the attributed low sensitivity of the C3H mouse strain to the loss of mechanical signals is not apparent at a young age and this trait therefore does not reflect a genetic regulation throughout the life span of this strain. These results highlight the significance of age in modulating the contribution of genetics in orchestrating bone’s response to unloading and that the skeletal unresponsiveness of young adult C3H mice to the loss of weight bearing is not genetically hard-wired.
Collapse
Affiliation(s)
- Bing Li
- Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Jeyantt Srinivas Sankaran
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Stefan Judex
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
8
|
Bloomfield SA, Martinez DA, Boudreaux RD, Mantri AV. Microgravity Stress: Bone and Connective Tissue. Compr Physiol 2016; 6:645-86. [PMID: 27065165 DOI: 10.1002/cphy.c130027] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The major alterations in bone and the dense connective tissues in humans and animals exposed to microgravity illustrate the dependency of these tissues' function on normal gravitational loading. Whether these alterations depend solely on the reduced mechanical loading of zero g or are compounded by fluid shifts, altered tissue blood flow, radiation exposure, and altered nutritional status is not yet well defined. Changes in the dense connective tissues and intervertebral disks are generally smaller in magnitude but occur more rapidly than those in mineralized bone with transitions to 0 g and during recovery once back to the loading provided by 1 g conditions. However, joint injuries are projected to occur much more often than the more catastrophic bone fracture during exploration class missions, so protecting the integrity of both tissues is important. This review focuses on the research performed over the last 20 years in humans and animals exposed to actual spaceflight, as well as on knowledge gained from pertinent ground-based models such as bed rest in humans and hindlimb unloading in rodents. Significant progress has been made in our understanding of the mechanisms for alterations in bone and connective tissues with exposure to microgravity, but intriguing questions remain to be solved, particularly with reference to biomedical risks associated with prolonged exploration missions.
Collapse
Affiliation(s)
- Susan A Bloomfield
- Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Daniel A Martinez
- Department of Mechanical Engineering, University of Houston, Houston, Texas, USA
| | - Ramon D Boudreaux
- Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Anita V Mantri
- Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA.,Health Science Center School of Medicine, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
9
|
Abstract
Bones adapt to accommodate the physical forces they experience through changes in architecture and mass. Stem cells differentiate into bone-forming osteoblasts, and mechanical stimulation is involved in this process. Various studies have applied controlled mechanical stimulation to stem cells and investigated the effects on osteogenic lineage commitment. These studies demonstrate that physical stimuli can induce osteogenic lineage commitment. Tension, fluid shear stress, substrate material properties, and cell shape are all factors that influence osteogenic differentiation. In particular, the level of tension is important. Also, rigid substrates with stiffness similar to collagenous bone induce osteogenic differentiation, while softer substrates induce other lineages. Finally, cells allowed to adhere over a larger area are able to differentiate towards the osteogenic lineage while cells adhering to a smaller area are restricted to the adipogenic lineage. Stem cells are able to sense their mechanical environments through various mechanosensors, including the cytoskeleton, focal adhesions, and primary cilia. The cytoskeleton provides a structural frame for the cell, and myosin interacts with actin to generate cytoskeletal tension, which is important for mechanically induced osteogenesis of stem cells. Adapter proteins link the cytoskeleton to integrins, which attach the cell to the substrate, forming a focal adhesion. A variety of signaling proteins are also associated with focal adhesions. Forces are transmitted to the substrate at these sites, and an intact focal adhesion is important for mechanically induced osteogenesis. The primary cilium is a single, immotile, antenna-like structure that extends from the cell into the extracellular space. It has emerged as an important signaling center, acting as a microdomain to facilitate biochemical signaling. Mechanotransduction is the process by which physical stimuli are converted into biochemical responses. When potential mechanosensors are disrupted, the activities of components of mechanotransduction pathways are also inhibited, preventing mechanically induced osteogenesis. Calcium, mitogen-activated protein kinase/extracellular signal-regulated kinase, Wnt, Yes-associated protein/transcriptional coactivator with PDZ-binding motif and RhoA/Rho kinase signaling are some of the mechanotransduction pathways proposed to be important. In this review, types of mechanical stimuli, mechanosensors, and key pathways involved in mechanically induced osteogenesis of stem cells are discussed.
Collapse
|
10
|
Nagaraja MP, Risin D. The current state of bone loss research: data from spaceflight and microgravity simulators. J Cell Biochem 2013; 114:1001-8. [PMID: 23150462 DOI: 10.1002/jcb.24454] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/01/2012] [Indexed: 11/11/2022]
Abstract
Bone loss is a well documented phenomenon occurring in humans both in short- and in long-term spaceflights. This phenomenon can be also reproduced on the ground in human and animals and also modeled in cell-based analogs. Since space flights are infrequent and expensive to study the biomedical effects of microgravity on the human body, much of the known pathology of bone loss comes from experimental studies. The most commonly used in vitro simulators of microgravity are clinostats while in vivo simulators include the bed rest studies in humans and hindlimb unloading experiments in animals. Despite the numerous reports that have documented bone loss in wide ranges in multiple crew members, the pathology remains a key concern and development of effective countermeasures is still a major task. Thus far, the offered modalities have not shown much success in preventing or alleviating bone loss in astronauts and cosmonauts. The objective of this review is to capture the most recent research on bone loss from spaceflights, bed rest and hindlimb unloading, and in vitro studies utilizing cellular models in clinostats. Additionally, this review offers projections on where the research has to focus to ensure the most rapid development of effective countermeasures.
Collapse
|
11
|
Wang YK, Chen CS. Cell adhesion and mechanical stimulation in the regulation of mesenchymal stem cell differentiation. J Cell Mol Med 2013; 17:823-32. [PMID: 23672518 PMCID: PMC3741348 DOI: 10.1111/jcmm.12061] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 03/01/2013] [Indexed: 12/15/2022] Open
Abstract
Stem cells have been shown to have the potential to provide a source of cells for applications to tissue engineering and organ repair. The mechanisms that regulate stem cell fate, however, mostly remain unclear. Mesenchymal stem cells (MSCs) are multipotent progenitor cells that are isolated from bone marrow and other adult tissues, and can be differentiated into multiple cell lineages, such as bone, cartilage, fat, muscles and neurons. Although previous studies have focused intensively on the effects of chemical signals that regulate MSC commitment, the effects of physical/mechanical cues of the microenvironment on MSC fate determination have long been neglected. However, several studies provided evidence that mechanical signals, both direct and indirect, played important roles in regulating a stem cell fate. In this review, we summarize a number of recent studies on how cell adhesion and mechanical cues influence the differentiation of MSCs into specific lineages. Understanding how chemical and mechanical cues in the microenvironment orchestrate stem cell differentiation may provide new insights into ways to improve our techniques in cell therapy and organ repair.
Collapse
Affiliation(s)
- Yang-Kao Wang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan.
| | | |
Collapse
|
12
|
Qin W, Sun L, Cao J, Peng Y, Collier L, Wu Y, Creasey G, Li J, Qin Y, Jarvis J, Bauman WA, Zaidi M, Cardozo C. The central nervous system (CNS)-independent anti-bone-resorptive activity of muscle contraction and the underlying molecular and cellular signatures. J Biol Chem 2013; 288:13511-21. [PMID: 23530032 DOI: 10.1074/jbc.m113.454892] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mechanisms by which muscle regulates bone are poorly understood. RESULTS Electrically stimulated muscle contraction reversed elevations in bone resorption and increased Wnt signaling in bone-derived cells after spinal cord transection. CONCLUSION Muscle contraction reduced resorption of unloaded bone independently of the CNS, through mechanical effects and, potentially, nonmechanical signals (e.g. myokines). SIGNIFICANCE The study provides new insights regarding muscle-bone interactions. Muscle and bone work as a functional unit. Cellular and molecular mechanisms underlying effects of muscle activity on bone mass are largely unknown. Spinal cord injury (SCI) causes muscle paralysis and extensive sublesional bone loss and disrupts neural connections between the central nervous system (CNS) and bone. Muscle contraction elicited by electrical stimulation (ES) of nerves partially protects against SCI-related bone loss. Thus, application of ES after SCI provides an opportunity to study the effects of muscle activity on bone and roles of the CNS in this interaction, as well as the underlying mechanisms. Using a rat model of SCI, the effects on bone of ES-induced muscle contraction were characterized. The SCI-mediated increase in serum C-terminal telopeptide of type I collagen (CTX) was completely reversed by ES. In ex vivo bone marrow cell cultures, SCI increased the number of osteoclasts and their expression of mRNA for several osteoclast differentiation markers, whereas ES significantly reduced these changes; SCI decreased osteoblast numbers, but increased expression in these cells of receptor activator of NF-κB ligand (RANKL) mRNA, whereas ES increased expression of osteoprotegerin (OPG) and the OPG/RANKL ratio. A microarray analysis revealed that ES partially reversed SCI-induced alterations in expression of genes involved in signaling through Wnt, FSH, parathyroid hormone (PTH), oxytocin, and calcineurin/nuclear factor of activated T-cells (NFAT) pathways. ES mitigated SCI-mediated increases in mRNA levels for the Wnt inhibitors DKK1, sFRP2, and sclerostin in ex vivo cultured osteoblasts. Our results demonstrate an anti-bone-resorptive activity of muscle contraction by ES that develops rapidly and is independent of the CNS. The pathways involved, particularly Wnt signaling, suggest future strategies to minimize bone loss after immobilization.
Collapse
Affiliation(s)
- Weiping Qin
- Departments of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Long RK, Nishida S, Kubota T, Wang Y, Sakata T, Elalieh HZ, Halloran BP, Bikle DD. Skeletal unloading-induced insulin-like growth factor 1 (IGF-1) nonresponsiveness is not shared by platelet-derived growth factor: the selective role of integrins in IGF-1 signaling. J Bone Miner Res 2011; 26:2948-58. [PMID: 21932337 PMCID: PMC3222734 DOI: 10.1002/jbmr.511] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Integrin receptors bind extracellular matrix proteins, and this link between the cell membrane and the surrounding matrix may translate skeletal loading to biologic activity in osteoprogenitor cells. The interaction between integrin and growth factor receptors allows for mechanically induced regulation of growth factor signaling. Skeletal unloading leads to decreased bone formation and osteoblast proliferation that can be explained in part by a failure of insulin-like growth factor 1 (IGF-1) to activate its signaling pathways in unloaded bone. The aim of this study is to determine whether unloading-induced resistance is specific for IGF-1 or common to other skeletal growth factors, and to examine the regulatory role of integrins in IGF-1 signaling. Bone marrow osteoprogenitor (BMOp) cells were isolated from control or hindlimb suspended rats. Unloaded BMOp cells treated with IGF-1 failed to respond with increased proliferation, receptor phosphorylation, or signaling activation in the setting of intact ligand binding, whereas the platelet-derived growth factor (PDGF) response was fully intact. Pretreatment of control BMOp cells with an integrin inhibitor, echistatin, failed to disrupt PDGF signaling but blocked IGF-1 signaling. Recovery of IGF-1 signaling in unloaded BMOp cells followed the recovery of marked reduction in integrin expression induced by skeletal unloading. Selective targeting of integrin subunits with siRNA oligonucleotides revealed that integrin β1 and β3 are required for normal IGF-1 receptor phosphorylation. We conclude that integrins, in particular integrin β3, are regulators of IGF-1, but not PDGF, signaling in osteoblasts, suggesting that PDGF could be considered for investigation in prevention and/or treatment of bone loss during immobilization and other forms of skeletal unloading.
Collapse
Affiliation(s)
- Roger K Long
- Department of Medicine, University of California, Davis, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Hoey DA, Kelly DJ, Jacobs CR. A role for the primary cilium in paracrine signaling between mechanically stimulated osteocytes and mesenchymal stem cells. Biochem Biophys Res Commun 2011; 412:182-7. [PMID: 21810408 DOI: 10.1016/j.bbrc.2011.07.072] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 07/19/2011] [Indexed: 01/07/2023]
Abstract
Bone turnover is a mechanically regulated process, coordinated in part by the network of mechanosensitive osteocytes residing within the tissue. The recruitment and bone forming activity of the mesenchymal derived osteoblast is determined by numerous factors including mechanical loading. It is therefore somewhat surprising that although mechanically regulated signaling between the coordinating osteocytes and mesenchymal stem cells (MSCs) should exist, to date it has not been directly demonstrated. In this study, conditioned media from mechanically stimulated osteocytes (MLO-Y4 cell line) was collected and added to MSCs (C3H10T1/2 cell line). The addition of mechanically stimulated osteocyte conditioned media resulted in a significant upregulation of the osteogenic genes OPN and COX-2 in MSCs compared to statically cultured conditioned media, demonstrating a novel paracrine signaling mechanism between the two cell types. The same mechanically conditioned media did not alter gene expression in osteoblasts (MC3T3 cell line), and mechanically stimulated osteoblast conditioned media did not alter gene expression in MSCs demonstrating that this signaling is unique to osteocytes and MSCs. Finally, the upregulation in osteogenic genes in MSCs was not observed if primary cilia formation was inhibited prior to mechanical stimulation of the osteocyte. In summary, the results of this study indicate that soluble factors secreted by osteocytes in response to mechanical stimulation can enhance osteogenic gene expression in MSCs demonstrating a novel, unique signaling mechanism and introduces a role for the primary cilium in flow mediated paracrine signaling in bone thereby highlighting the cilium as a potential target for therapeutics aimed at enhancing bone formation.
Collapse
Affiliation(s)
- David A Hoey
- Department of Biomedical Engineering, Columbia University in the City of New York, NY, USA.
| | | | | |
Collapse
|
15
|
Marie PJ, Kaabeche K. PPAR Gamma Activity and Control of Bone Mass in Skeletal Unloading. PPAR Res 2011; 2006:64807. [PMID: 17259667 PMCID: PMC1679962 DOI: 10.1155/ppar/2006/64807] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 06/20/2006] [Accepted: 07/05/2006] [Indexed: 11/18/2022] Open
Abstract
Bone loss occuring with unloading is associated with decreased
osteoblastogenesis and increased bone marrow adipogenesis, resulting
in bone loss and decreased bone formation. Here, we review the present knowledge on the role of PPARγ in the control of osteoblastogenesis and bone mass in skeletal unloading. We showed that PPARγ positively promotes adipogenesis and negatively regulates osteoblast differentiation of bone marrow stromal cells in unloading, resulting in bone loss. Manipulation of PPARγ2 expression by exogenous TGF-β2 inhibits the exaggerated adipogenesis and corrects the balance between osteoblastogenesis and adipogenesis induced by unloading, leading to prevention of bone loss. This shows that PPARγ plays an important role in the control of bone mass in unloaded bone. Moreover, this opens the possibility that manipulation of PPARγ may correct the balance between osteoblastogenesis and adipogenesis and prevent bone loss,
which may have potential implications in the treatment of bone loss in clinical
conditions.
Collapse
Affiliation(s)
- P. J. Marie
- Laboratory of Osteoblast Biology and Pathology, INSERM U606, 75475 Paris Cedex 10, France
- Faculty of Medicine, University of Paris 7, 75251 Paris Cedex 05, France
- *P. J. Marie:
| | - K. Kaabeche
- Laboratory of Osteoblast Biology and Pathology, INSERM U606, 75475 Paris Cedex 10, France
- Faculty of Medicine, University of Paris 7, 75251 Paris Cedex 05, France
| |
Collapse
|
16
|
Habold C, Momken I, Ouadi A, Bekaert V, Brasse D. Effect of prior treatment with resveratrol on density and structure of rat long bones under tail-suspension. J Bone Miner Metab 2011; 29:15-22. [PMID: 20458604 DOI: 10.1007/s00774-010-0187-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 04/01/2010] [Indexed: 01/26/2023]
Abstract
Physical inactivity during space flight or prolonged bed rest causes rapid and marked loss of bone mass in humans. Resveratrol, a red wine polyphenol that is currently under study for its therapeutic antioxidant properties, has been shown to significantly modulate biomarkers of bone metabolism, i.e., to promote osteoblast differentiation and to prevent bone loss induced by estrogen deficiency. However, there is no direct evidence supporting its inhibitory effect toward bone loss during physical inactivity. In the present study, effects of resveratrol on bone mineral density (BMD), bone mineral content, and bone structure were examined in the femora and tibiae of tail-suspended and unsuspended rats using X-ray micro-computed tomography (micro-CT). Rats were treated with 400 mg/kg/day of resveratrol for 45 days and half of them were suspended during the last 2 weeks of treatment. Suspension caused a decrease in tibial and femoral BMD and deterioration of trabecular and cortical bone. Bone deterioration during suspension was paralleled by increased bone marrow area, which could be caused by an increase in stromal cells with osteoclastogenic potential or in adipocytes. Resveratrol had a preventive effect against bone loss induced by hindlimb immobilization. In particular, trabecular bone in the proximal tibial metaphysis was totally preserved in rats treated with resveratrol before tail-suspension.
Collapse
Affiliation(s)
- Caroline Habold
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR 7178, Université de Strasbourg, 23 rue du Loess, Strasbourg Cedex 2, France.
| | | | | | | | | |
Collapse
|
17
|
Rissanen JP, Halleen JM. Models and screening assays for drug discovery in osteoporosis. Expert Opin Drug Discov 2010; 5:1163-74. [DOI: 10.1517/17460441.2010.532484] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Marinucci L, Balloni S, Becchetti E, Bistoni G, Calvi EM, Lumare E, Ederli F, Locci P. Effects of Hydroxyapatite and Biostite® on Osteogenic Induction of hMSC. Ann Biomed Eng 2010; 38:640-8. [DOI: 10.1007/s10439-009-9898-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 12/30/2009] [Indexed: 10/20/2022]
|
19
|
Chen M, Feng W, Cao H, Zou L, Chen C, Baatrup A, Nielsen AB, Li H, Kassem M, Zou X, Bünger C. A traditional Chinese medicine formula extracts stimulate proliferation and inhibit mineralization of human mesenchymal stem cells in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2009; 125:75-82. [PMID: 19549563 DOI: 10.1016/j.jep.2009.06.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 05/04/2009] [Accepted: 06/14/2009] [Indexed: 05/28/2023]
Abstract
AIM OF THE STUDY To investigate the effects of a traditional Chinese medicine (TCM) formula extract, named as ZD-I, on the proliferation and osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro. MATERIALS AND METHODS When hMSCs cultivated in the basal medium with ZD-I, cell viability was assessed by MTT assay and cellular proliferation was assessed by SYBR green I assay. The effects of ZD-I on osteogenic differentiation of hMSCs were assessed by alkaline phosphatase (ALP) activity, mineralization assay and real-time RT-PCR. RESULTS ZD-I (0.78-100 microg/ml) was non-cytotoxic. The 50% inhibitory concentration (IC50) of hMSCs was 200 microg/ml. ZD-I (0.78-50 microg/ml) stimulated the proliferation of hMSCs. ZD-I did not change ALP activity of hMSCs cultivated in osteogenic medium in the early stage (4 and 7 days), but ZD-I inhibited the mineralization of hMSCs through down-regulation of several osteogenic markers (e.g. osteocalcin, bone morphogenetic protein 2 and osteopontin) in the late stage. CONCLUSIONS ZD-I stimulate cellular proliferation and decrease the bone mineral deposition of hMSCs. These results suggest ZD-I may play an important therapeutic role in osteoarthritic patients by improving proliferative capacity of hMSCs and inhibiting the mineralization of hMSCs.
Collapse
Affiliation(s)
- Muwan Chen
- Orthopaedic Research Lab, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Jekir MG, Donahue HJ. Gap junctions and osteoblast-like cell gene expression in response to fluid flow. J Biomech Eng 2009; 131:011005. [PMID: 19045921 DOI: 10.1115/1.3005201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bone formation occurs in vivo in response to mechanical stimuli, but the signaling pathways involved remain unclear. The ability of bone cells to communicate with each other in the presence of an applied load may influence the overall osteogenic response. The goal of this research was to determine whether inhibiting cell-to-cell gap junctional communication between bone-forming cells would affect the ensemble cell response to an applied mechanical stimulus in vitro. In this study, we investigated the effects of controlled oscillatory fluid flow (OFF) on osteoblastic cells in the presence and the absence of a gap-junction blocker. MC3T3-E1 Clone 14 cells in a monolayer were exposed to 2 h of OFF at a rate sufficient to create a shear stress of 20 dyne/cm(2) at the cell surface, and changes in steady-state mRNA levels for a number of key proteins known to be involved in osteogenesis were measured. Of the five proteins investigated, mRNA levels for osteopontin (OPN) and osteocalcin were found to be significantly increased 24 h postflow. These experiments were repeated in the presence of 18 beta-glycyrrhetinic acid (BGA), a known gap-junction blocker, to determine whether gap-junction intercellular communication is necessary for this response. We found that the increase in OPN mRNA levels is not observed in the presence of BGA, suggesting that gap junctions are involved in the signaling process. Interestingly, enzyme linked immunosorbent assay data showed that levels of secreted OPN protein increased 48 h postflow and that this increase was unaffected by the presence of intact gap junctions.
Collapse
Affiliation(s)
- Michael G Jekir
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
21
|
Riddle RC, Donahue HJ. From streaming-potentials to shear stress: 25 years of bone cell mechanotransduction. J Orthop Res 2009; 27:143-9. [PMID: 18683882 DOI: 10.1002/jor.20723] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mechanical loads are vital regulators of skeletal mass and architecture as evidenced by the increase in bone formation following the addition of exogenous loads and loss of bone mass following their removal. While our understanding of the molecular mechanisms by which bone cells perceive changes in their mechanical environment has increased rapidly in recent years, much remains to be learned. Here, we outline the effects of interstitial fluid flow, a potent biophysical signal induced by the deformation of skeletal tissue in response to applied loads, on bone cell behavior. We focus on the molecular mechanisms by which bone cells are hypothesized to perceive interstitial fluid flow, the cell signaling cascades activated by fluid flow, and the use of this signal in tissue engineering protocols.
Collapse
Affiliation(s)
- Ryan C Riddle
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
22
|
Dufour C, Holy X, Marie PJ. Transforming growth factor-beta prevents osteoblast apoptosis induced by skeletal unloading via PI3K/Akt, Bcl-2, and phospho-Bad signaling. Am J Physiol Endocrinol Metab 2008; 294:E794-801. [PMID: 18378961 DOI: 10.1152/ajpendo.00791.2007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Loss of mechanical loading induces rapid bone loss resulting from reduced osteoblastogenesis and decreased bone formation. The signaling mechanisms involved in this deleterious effect on skeletal metabolism remain poorly understood. We have previously shown that hindlimb suspension in rats increases osteoblast apoptosis associated with decreased phosphatidylinositol 3-kinase (PI3K) signaling. In this study, we investigated whether transforming growth factor (TGF)-beta2 may prevent the altered signaling and osteoblast apoptosis induced by skeletal unloading in vivo. Hindlimb suspension-induced decreased bone volume was associated with reduced alpha(5)beta(1)-integrin protein levels and PI3K/Akt signaling in unloaded bone. Continuous administration of TGF-beta2 using osmotic minipumps prevented the decreased alpha(5)beta(1)-integrin expression and the reduced PI3K/Akt signaling in unloaded bone, resulting in the prevention of osteoblast apoptosis. We also show that TGF-beta2 prevented the decreased Bcl-2 levels induced by unloading, which suggests that TGF-beta2 targets Bcl-2 via PI3K/Akt to prevent osteoblast apoptosis in unloaded bone. Furthermore, we show that TGF-beta2 prevented the decrease in phosphorylated Bad, the inactive form of the proapoptotic protein Bad, induced by unloading. These results identify a protective role for TGF-beta2 in osteoblast apoptosis induced by mechanical unloading via the alpha(5)beta(1)/PI3K/Akt signaling cascade and downstream Bcl-2 and phospho-Bad survival proteins. We thus propose a novel role for TGF-beta2 in protection from unloading-induced apoptosis in vivo.
Collapse
Affiliation(s)
- Cécilie Dufour
- Unit 606 Institut National de la Santé et de la Recherche Médicale, Paris, France
| | | | | |
Collapse
|
23
|
Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen MR, Alam I, Mantila SM, Gluhak-Heinrich J, Bellido TM, Harris SE, Turner CH. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem 2007; 283:5866-75. [PMID: 18089564 DOI: 10.1074/jbc.m705092200] [Citation(s) in RCA: 955] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sclerostin, the protein product of the Sost gene, is a potent inhibitor of bone formation. Among bone cells, sclerostin is found nearly exclusively in the osteocytes, the cell type that historically has been implicated in sensing and initiating mechanical signaling. The recent discovery of the antagonistic effects of sclerostin on Lrp5 receptor signaling, a crucial mediator of skeletal mechanotransduction, provides a potential mechanism for the osteocytes to control mechanotransduction, by adjusting their sclerostin (Wnt inhibitory) signal output to modulate Wnt signaling in the effector cell population. We investigated the mechanoregulation of Sost and sclerostin under enhanced (ulnar loading) and reduced (hindlimb unloading) loading conditions. Sost transcripts and sclerostin protein levels were dramatically reduced by ulnar loading. Portions of the ulnar cortex receiving a greater strain stimulus were associated with a greater reduction in Sost staining intensity and sclerostin-positive osteocytes (revealed via in situ hybridization and immunohistochemistry, respectively) than were lower strain portions of the tissue. Hindlimb unloading yielded a significant increase in Sost expression in the tibia. Modulation of sclerostin levels appears to be a finely tuned mechanism by which osteocytes coordinate regional and local osteogenesis in response to increased mechanical stimulation, perhaps via releasing the local inhibition of Wnt/Lrp5 signaling.
Collapse
Affiliation(s)
- Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Globus RK. Extracellular Matrix and Integrin Interactions in the Skeletal Responses to Mechanical Loading and Unloading. Clin Rev Bone Miner Metab 2007. [DOI: 10.1007/s12018-008-9013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
25
|
Dai ZQ, Wang R, Ling SK, Wan YM, Li YH. Simulated microgravity inhibits the proliferation and osteogenesis of rat bone marrow mesenchymal stem cells. Cell Prolif 2007; 40:671-84. [PMID: 17877609 PMCID: PMC6496371 DOI: 10.1111/j.1365-2184.2007.00461.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Microgravity is known to affect the differentiation of bone marrow mesenchymal stem cells (BMSCs). However, a few controversial findings have recently been reported with respect to the effects of microgravity on BMSC proliferation. Thus, we investigated the effects of simulated microgravity on rat BMSC (rBMSC) proliferation and their osteogeneic potential. MATERIALS AND METHODS rBMSCs isolated from marrow using our established effective method, based on erythrocyte lysis, were identified by their surface markers and their proliferation characteristics under normal conditions. Then, they were cultured in a clinostat to simulate microgravity, with or without growth factors, and in osteogenic medium. Subsequently, proliferation and cell cycle parameters were assessed using methylene blue staining and flow cytometry, respectively; gene expression was determined using Western blotting and microarray analysis. RESULTS Simulated microgravity inhibited population growth of the rBMSCs, cells being arrested in the G(0)/G(1) phase of cell cycle. Growth factors, such as insulin-like growth factor-I, epidermal growth factor and basic fibroblastic growth factor, markedly stimulated rBMSC proliferation in normal gravity, but had only a slight effect in simulated microgravity. Akt and extracellular signal-related kinase 1/2 phosphorylation levels and the expression of core-binding factor alpha1 decreased after 3 days of clinorotation culture. Microarray and gene ontology analyses further confirmed that rBMSC proliferation and osteogenesis decreased under simulated microgravity. CONCLUSIONS The above data suggest that simulated microgravity inhibits population growth of rBMSCs and their differentiation towards osteoblasts. These changes may be responsible for some of the physiological changes noted during spaceflight.
Collapse
Affiliation(s)
- Z Q Dai
- China Astronaut Research and Training Center, Laboratory of Space Cell and Molecular Biology, Beijing, China
| | | | | | | | | |
Collapse
|
26
|
Riddle RC, Taylor AF, Rogers JR, Donahue HJ. ATP release mediates fluid flow-induced proliferation of human bone marrow stromal cells. J Bone Miner Res 2007; 22:589-600. [PMID: 17243863 DOI: 10.1359/jbmr.070113] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
UNLABELLED Oscillatory fluid flow induced the vesicular release of ATP from human BMSCs that directly contributes to the induction of BMSC proliferation. Degrading extracellular nucleotides prevents fluid flow-induced increases in intracellular calcium concentration, the activation of calcineurin, and the nuclear translocation of NFAT. INTRODUCTION Regulation of bone cell activity by autocrine/paracrine factors is a well-established mechanism by which skeletal homeostasis is regulated by mechanical signals. The release of extracellular nucleotides in particular has been shown to induce many of the responses thought to be necessary for load-induced bone formation. In these studies, we examined the effect of oscillatory fluid flow on the release of ATP from bone marrow stromal cells (BMSCs) and the effect of ATP release on BMSC proliferation and intracellular calcium signaling pathways. MATERIALS AND METHODS BMSCs were exposed to oscillatory fluid flow, and the concentration of ATP in conditioned media samples was determined using a luciferin:luciferase-based reaction. Western blot analysis was used to examine the expression of purinergic receptors. Using pharmacological antagonists of gap junction hemichannels and vesicular trafficking, we studied the mechanism of ATP release from BMSCs. Apyrase was used to study the effect of extracellular nucleotides on intracellular calcium concentration, calcineurin activity, and nuclear factor of activated T cells (NFAT) nuclear translocation. RESULTS AND CONCLUSIONS Fluid flow exposure induced the flow rate-dependent release of ATP from BMSCs that was attenuated by treatment with monensin and N-ethylmaleimide, suggesting a vesicular mechanism. Treating BMSCs with ATP, but not other nucleotides, increased cellular proliferation. Moreover, extracellular ATP was a prerequisite for fluid flow-induced increases in intracellular calcium concentration, activation of calcineurin, the nuclear translocation of NFATc1, and proliferation. These data indicate that ATP regulates not only osteoblastic and osteocytic cell behavior but also that of mesenchymal precursors and support our hypothesis that similar mechanotransduction mechanisms are activated by fluid flow in these cell types.
Collapse
Affiliation(s)
- Ryan C Riddle
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Pennsylvania State University College of Medicine, Milton S Hershey Medical Center, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
27
|
Dufour C, Holy X, Marie PJ. Skeletal unloading induces osteoblast apoptosis and targets α5β1-PI3K-Bcl-2 signaling in rat bone. Exp Cell Res 2007; 313:394-403. [PMID: 17123509 DOI: 10.1016/j.yexcr.2006.10.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 10/23/2006] [Accepted: 10/23/2006] [Indexed: 11/16/2022]
Abstract
The mechanisms underlying the altered osteoblastogenesis and bone loss in response to disuse are incompletely understood. Using the rat tail suspension model, we studied the effect of skeletal unloading on osteoblast and osteocyte apoptosis. Tail suspension for 2 to 7 days decreased tibial bone mass and induced early apoptotic loss of osteoblasts and delayed apoptotic loss of osteocytes. Surrenal gland weight and plasma corticosterone levels did not differ in loaded and unloaded rats at any time point, indicating that osteoblast/osteocyte apoptosis occurred independently of endogenous glucocorticoids. The mechanistic basis for the disuse-induced osteoblast/osteocyte apoptosis was examined. We found that alpha5beta1 integrin and phosphorylated phosphatidyl-inositol-3 kinase (p-PI3K) protein levels were transiently decreased in unloaded metaphyseal long bone compared to loaded bones. In contrast, p-FAK and p-ERK p42/44 levels were not significantly altered. Interestingly, the reduced p-PI3K levels in unloaded long bone was associated with decreased levels of the survival protein Bcl-2 with unaltered Bax levels, causing increased Bax/Bcl-2 levels. The results indicate that skeletal unloading in rats induces a glucocorticoid-independent, immediate increase in osteoblast apoptosis associated with decreased alpha5beta1-PI3K-Bcl-2 survival pathway in rat bone, which may contribute to the altered osteoblastogenesis and osteopenia induced by unloading.
Collapse
Affiliation(s)
- C Dufour
- Unit 606 INSERM, Laboratory of Osteoblast Biology and Pathology, University Paris 7, France
| | | | | |
Collapse
|
28
|
Perrien DS, Akel NS, Dupont-Versteegden EE, Skinner RA, Siegel ER, Suva LJ, Gaddy D. Aging alters the skeletal response to disuse in the rat. Am J Physiol Regul Integr Comp Physiol 2006; 292:R988-96. [PMID: 17068163 DOI: 10.1152/ajpregu.00302.2006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disuse has been shown to cause a rapid and dramatic loss of skeletal mass and strength in the load-bearing bones of young and mature animals and humans. However, little is known about the skeletal effects of disuse in aged mammals. The present study was designed to determine whether the skeletal effects of disuse are maintained with extreme age. Fischer 344/Brown Norway male rats (6 and 32 mo old) were hindlimb suspended (HS) or housed individually for 2 wk. Trabecular volume and microarchitecture in the proximal tibia were significantly decreased by HS only in young rats. HS significantly reduced cortical bone mineral density and increased cortical porosity only in old rats by inducing new pore formation. Cortical pore diameter was also increased in old rats, regardless of loading condition. Ex vivo osteogenic and adipogenic cultures established from each group demonstrated that age and HS decreased osteoblastogenesis. Age, but not HS, decreased sensitivity to endogenous bone morphogenetic protein stimulation, as measured by treatment with exogenous Noggin. Adipocyte development increased with age, whereas HS suppressed sensitivity to peroxisome proliferator-activated receptor-gamma-induced differentiation. Serum insulin-like growth factor I levels were reduced with HS in young rats and with age in control and HS rats. These results suggest that the site of bone loss due to disuse is altered with age and that the loss of osteogenic potential with disuse in the old rats may be due to the combined effects of decreased insulin-like growth factor I levels and sensitivity, as well as diminished bone morphogenetic protein production.
Collapse
Affiliation(s)
- Daniel S Perrien
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, 4301 West Markham St., Slot 505, Little Rock, AR 72205, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Tjabringa GS, Vezeridis PS, Zandieh-Doulabi B, Helder MN, Wuisman PIJM, Klein-Nulend J. Polyamines Modulate Nitric Oxide Production andCox-2Gene Expression in Response to Mechanical Loading in Human Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells 2006; 24:2262-9. [PMID: 16794268 DOI: 10.1634/stemcells.2005-0625] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
For bone tissue engineering, it is important that mesenchymal stem cells (MSCs) display a bone cell-like response to mechanical loading. We have shown earlier that this response includes increased nitric oxide (NO) production and cyclooxygenase-2 (COX-2) gene expression, both of which are intimately involved in mechanical adaptation of bone. COX-2 gene expression is likely regulated by polyamines, which are organic cations implicated in cell proliferation and differentiation. This has led to the hypothesis that polyamines may play a role in the response of adipose tissue-derived MSCs (AT-MSCs) to mechanical loading. The aim of this study was to investigate whether genes involved in polyamine metabolism are regulated by mechanical loading and to study whether polyamines modulate mechanical loading-induced NO production and COX-2 gene expression in human AT-MSCs. Human AT-MSCs displayed a bone cell-like response to mechanical loading applied by pulsating fluid flow (PFF), as demonstrated by increased NO production and increased gene expression of COX-2. Furthermore, PFF increased gene expression of spermidine/spermine N (1)-acetyltransferase, which is involved in polyamine catabolism, suggesting that mechanical loading modulates polyamine levels. Finally, the polyamine spermine was shown to inhibit both PFF-induced NO production and COX-2 gene expression, suggesting that polyamines modulate the response of human AT-MSCs to mechanical loading. In conclusion, this is the first study implicating polyamines in the response of human AT-MSCs to mechanical loading, creating opportunities for the use of polyamines in tissue engineering approaches targeting skeletal defects.
Collapse
Affiliation(s)
- Geuranne S Tjabringa
- Department of Oral Cell Biology, Academic Center of Dentistry Amsterdam, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
30
|
Christiansen BA, Silva MJ. The Effect of Varying Magnitudes of Whole-Body Vibration on Several Skeletal Sites in Mice. Ann Biomed Eng 2006; 34:1149-56. [PMID: 16786394 DOI: 10.1007/s10439-006-9133-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 05/04/2006] [Indexed: 12/26/2022]
Abstract
It has been reported that whole-body vibration (WBV) is anabolic to trabecular bone in animal models and humans. It is likely that this anabolic response does not occur uniformly throughout the entire body. Two factors that may affect the observed anabolic response are vibration magnitude and skeletal site of interest. In this study, mice were loaded with WBV of varying magnitudes. After five weeks of loading, bone marrow was flushed from tibias in order to quantify osteoprogenitor cells. Staining with alizarin red (an indicator of mineralization) showed a significant decrease in percent stained area in the 0.3 g loaded group compared to the control group and the 1.0 g group. MicroCT analysis was performed at five skeletal sites: the proximal tibial metaphysis, femoral condyles, distal femoral metaphysis, proximal femur, and L5 vertebral body. Increasing magnitudes of WBV were associated with a non-dose-dependent increase in trabecular bone volume (BV/TV) at the proximal tibial metaphysis, although other sites were unresponsive. There were statistically significant increases in BV/TV in the 0.1 g group (32% increase) and 1.0 g group (43% increase) compared to control (p < 0.05). The 0.1 g and 1.0 g groups also had higher BV/TV than the 0.3 g loaded group. If this non-dose-dependent phenomenon is verified by future studies, it suggests that a range of magnitudes should be examined for each application of WBV.
Collapse
Affiliation(s)
- Blaine A Christiansen
- Department of Orthopaedic Surgery, University in St. Louis, 1 Barnes-Jewish Hospital Plaza Campus Box 8233, St. Louis, MO 63110, USA.
| | | |
Collapse
|
31
|
Ho ML, Tsai TN, Chang JK, Shao TS, Jeng YR, Hsu C. Down-regulation of N-methyl D-aspartate receptor in rat-modeled disuse osteopenia. Osteoporos Int 2005; 16:1780-8. [PMID: 15997422 DOI: 10.1007/s00198-005-1928-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Accepted: 04/14/2005] [Indexed: 10/25/2022]
Abstract
Lack of mechanical stress may result in osteoporosis; however, the underlying mechanisms of disuse osteoporosis remain unclear. It has been indicated that mechanical loading causes extracellular glutamate accumulation in osteoblasts. We hypothesized that the glutamate receptor mediation on bone cells might also be involved in mechanically stimulated osteogenesis. In this study, we investigated the changes of bone formation and the expressions of osteogenic genes and N-methyl D-aspartate (NMDA) receptors, the major glutamate receptors, in disused bones. Rat modeled disuse osteopenia in hind limbs was induced by a 3-week tail suspension in Sprague-Dawley rats. Bone mineral density and trabecular bone volume of distal femurs were measured to verify the osteopenia of disused bones. The mRNA expressions of cbfa1/Runx2, type I collagen, alkaline phosphatase (ALP) and osteocalcin (OC) in bones were measured as osteogenic markers. The influences of mechanical unloading on the expressions of NMDA receptors (NR1 and NR2D) in bones were also examined. The effects of NMDA mediation on osteogenesis were tested by a treatment of MK-801, a non-competitive NMDA receptor antagonist, in cultured osteoblasts and bone marrow stroma cells. Our result showed that mRNA expressions of cbfa1/Runx2, type I collagen, ALP and OC were significantly decreased in disused bones. The mRNA and protein expressions of NR1 and NR2D were significantly decreased in disused bones; furthermore, immunolocalization of both receptors showed decreases in osteoblasts, but not in osteoclasts. The results from the in vitro study showed that MK-801 inhibited mRNA expression of cbfa1/Runx2 in bone marrow stroma cells and also inhibited those of collagen type I, ALP and OC of osteoblasts in a dose-dependent manner. These results suggest that NMDA receptor mediation may play an important role in transmitting mechanical loading in bones, and decreases of the expressions of NMDA receptors in disused bones, especially in osteoblasts, may contribute to the decrease of osteogenesis.
Collapse
Affiliation(s)
- Mei-Ling Ho
- Department of Physiology, Kaohsiung Medical University, No. 100 Shih-Chuan 1st Road, 807, Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Riddle RC, Taylor AF, Genetos DC, Donahue HJ. MAP kinase and calcium signaling mediate fluid flow-induced human mesenchymal stem cell proliferation. Am J Physiol Cell Physiol 2005; 290:C776-84. [PMID: 16267109 DOI: 10.1152/ajpcell.00082.2005] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanical signals are important regulators of skeletal homeostasis, and strain-induced oscillatory fluid flow is a potent mechanical stimulus. Although the mechanisms by which osteoblasts and osteocytes respond to fluid flow are being elucidated, little is known about the mechanisms by which bone marrow-derived mesenchymal stem cells respond to such stimuli. Here we show that the intracellular signaling cascades activated in human mesenchymal stem cells by fluid flow are similar to those activated in osteoblastic cells. Oscillatory fluid flow inducing shear stresses of 5, 10, and 20 dyn/cm(2) triggered rapid, flow rate-dependent increases in intracellular calcium that pharmacological studies suggest are inositol trisphosphate mediated. The application of fluid flow also induced the phosphorylation of extracellular signal-regulated kinase-1 and -2 as well as the activation of the calcium-sensitive protein phosphatase calcineurin in mesenchymal stem cells. Activation of these signaling pathways combined to induce a robust increase in cellular proliferation. These data suggest that mechanically induced fluid flow regulates not only osteoblastic behavior but also that of mesenchymal precursors, implying that the observed osteogenic response to mechanical loading may be mediated by alterations in the cellular behavior of multiple members of the osteoblast lineage, perhaps by a common signaling pathway.
Collapse
Affiliation(s)
- Ryan C Riddle
- Department of Orthopaedics and Rehabilitation, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Ctr., Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
33
|
Sakuma-Zenke M, Sakai A, Nakayamada S, Kunugita N, Tabata T, Uchida S, Tanaka S, Mori T, Nakai K, Tanaka Y, Nakamura T. Reduced expression of platelet endothelial cell adhesion molecule-1 in bone marrow cells in mice after skeletal unloading. J Bone Miner Res 2005; 20:1002-10. [PMID: 15883641 DOI: 10.1359/jbmr.050102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2004] [Revised: 08/26/2004] [Accepted: 12/29/2004] [Indexed: 11/18/2022]
Abstract
UNLABELLED One week of tail suspension significantly decreased the expression of PECAM-1 in mouse tibial bone marrow cells but not those of a number of other vascular factors. Anti-PECAM-1 antibody suppressed both ALP+ CFU-f formation and ALP production under co-culture of the osteoblastic cell line and the PECAM-1+ endothelial cell line. This study suggests that the reduced ALP activity after skeletal unloading is related to downregulation of PECAM-1 expression in bone marrow cells in mice. INTRODUCTION Vascular factors play a role in bone development and regeneration. We tested the hypothesis that skeletal unloading reduces osteogenic potential by inhibiting the molecules related to angiogenesis and/or vasculogenesis in bone marrow cells. MATERIALS AND METHODS Eight-week-old male mice were assigned to three groups after acclimatization for 1 week: ground control (GC), tail suspension (TS), and reloading after 7-day TS (RL). Bilateral tibial and humeral samples were used for analyses. MC3T3-E1, a mouse osteoblastic cell line, and EOMA and ISOS-1, mouse endothelial cell lines, were also used. RESULTS Flow cytometric analysis revealed that 7-day TS significantly decreased the expression of platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) in tibial bone marrow cells, but not those of angiopoietin-1, angiopoietin-2, Flk-1 (vascular endothelial growth factor receptor-2), and vascular endothelial cadherin. The expression of PECAM-1 in tibial marrow cells was reduced at day 3 of TS to 80% and still showed significantly low levels at day 7 of TS to 72% of that at the respective days of GC. This decreased expression of PECAM-1 after 7-day TS showed the GC level at 5-day reloading after 7-day TS. However, the expression of PECAM-1 in humeral marrow cells (internal bone marrow control) after TS and RL remained unchanged and equivalent to that of GC. The expression level of PECAM-1 mRNA was significantly lower at day 7 of TS to 62% of that in GC. Double labeling analyses revealed that PECAM-1+ cells mostly consisted of endothelial cells and partially of granulocytes. In bone marrow cell cultures, the formation of alkaline phosphatase (ALP)+ colony forming units-fibroblastic was significantly reduced in the presence of anti-PECAM-1 antibody in the medium compared with the presence of immunoglobulin G (0.025 times as much as ALP production with immunoglobulin G). ALP production by cultured MC3T3-E1 was enhanced in combination with PECAM-1+ EOMA (1.8 times as much as ALP production by MC3T3-E1 alone), but not in combination with PECAM-1- ISOS-1. Anti-PECAM-1 antibody inhibited the increase in ALP production under co-culture with EOMA. CONCLUSIONS Our data show that the reduced ALP activity after skeletal unloading is closely correlated with reduced expression of PECAM-1 in bone marrow cells. We speculate that the loss of osteogenic potential after skeletal unloading is caused by the suppression of PECAM-1 signaling on endothelial cellular surface.
Collapse
Affiliation(s)
- Miyuki Sakuma-Zenke
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Iseigaoka, Kitakyushu, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Basso N, Bellows CG, Heersche JNM. Effect of simulated weightlessness on osteoprogenitor cell number and proliferation in young and adult rats. Bone 2005; 36:173-83. [PMID: 15664015 DOI: 10.1016/j.bone.2004.09.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Revised: 09/22/2004] [Accepted: 09/24/2004] [Indexed: 11/29/2022]
Abstract
Experiments with rats flown in space or hind limb unloaded (HU) indicate that bone loss in both conditions is associated with a decrease in bone volume and osteoblast surface in cancellous and cortical bone. We hypothesize that the decrease in osteoblastic bone formation and osteoblast surface is related to a decrease in the number of osteoprogenitors and/or decreased proliferation of their progeny. We tested this hypothesis by evaluating the effect of 14 days of HU on the number of osteoprogenitors (osteoblast colony forming units; CFU-O), fibroblastic colony forming units (CFU-F), and alkaline phosphatase-positive CFU (CFU-AP) in cell populations derived from the proximal femur (unloaded) and the proximal humerus (normally loaded) in 6-week-old and 6-month-old rats. To confirm the effect of unloading on bone volume and structure, static histomorphometric parameters were measured in the proximal tibial metaphysis. Effects of HU on proliferation of osteoprogenitors were evaluated by measuring the size of CFU-O. HU did not affect the total number of progenitors (CFU-F) in young or adult rats in any of the cell populations. In femoral populations of young rats, HU decreased CFU-O by 71.0% and mean colony size was reduced by 20%. HU decreased CFU-AP by 31.3%. As expected, no changes in CFU-O or CFU-AP were seen in cell populations from the humerus. In femoral cell populations of adult rats, HU decreased CFU-O and CFU-AP by 16.6% and 36.6%, respectively. Again, no effects were seen in cell populations from the humerus. In 6-week-old rats, there was a greater decrease in bone volume, osteoblast number, and osteoblast surface in the proximal tibial metaphysis than that observed in adult rats. Both trabecular thickness and trabecular number were decreased in young rats but remained unaffected in adults. Neither osteoclast number nor surface was affected by unloading. Our results show that the HU-induced decrease in the number of osteoprogenitors observed in vitro parallels the effects of HU on bone volume and osteoblast number in young and old rats in vivo, suggesting that the two may be interdependent. HU also reduced CFU-O colony size in femoral populations indicating a diminished proliferative capacity of osteoblastic colonies.
Collapse
Affiliation(s)
- N Basso
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada M5G 1G6.
| | | | | |
Collapse
|
35
|
Li YJ, Batra NN, You L, Meier SC, Coe IA, Yellowley CE, Jacobs CR. Oscillatory fluid flow affects human marrow stromal cell proliferation and differentiation. J Orthop Res 2004; 22:1283-9. [PMID: 15475210 DOI: 10.1016/j.orthres.2004.04.002] [Citation(s) in RCA: 179] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mechanical loading is an important regulator of bone formation and bone loss. Decreased osteoblast number and function are important cellular mechanisms by which mechanical disuse leads to decreased bone formation. Decreased osteoblast number may be a result of decreased osteoprogenitor proliferation, differentiation, or both. However, the effects of cellular level physical signals on osteoprogenitors are not well understood. In this study, we examined the effects of loading induced oscillatory fluid flow (OFF), a potent regulator of osteoblastic cell function, on marrow stromal cells (MSCs). MSCs subjected to OFF exhibited increased intracellular Ca2+ mobilization. In addition, MSCs exhibited increased proliferation and increased mRNA levels for osteopontin and osteocalcin genes. Collagen I and core binding factor 1 mRNA levels did not change. MSCs subjected to OFF also exhibited decreased alkaline phosphatase activity. These results suggest that MSCs are mechanosensitive and that Ca2+ may play a role in the signaling pathway.
Collapse
Affiliation(s)
- Ying Jun Li
- Palo Alto Veterans Administration Medical Center, CA 94304, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
In recent years, stem cells have shown significant promise for their potential to provide a source of undifferentiated progenitor cells for therapeutic applications in tissue or organ repair. Significant questions still remain, however, as to the genetic and epigenetic signals that regulate the fate of stem cells. It is now well accepted that the micro-environment of the stem cell can have a significant influence on its differentiation and phenotypic expression. Although emphasis has been placed in previous work on the role of soluble mediators such as growth factors and cytokines on stem cell differentiation, there is now significant evidence, both direct and indirect, that mechanical signals may also regulate stem cell fate. We review a number of in vivo and in vitro studies that have provided evidence that mechanical factors have the ability to influence the differentiation of a number of cells that have been classified as either precursor, progenitor, or stem cells. Taken together, these studies show that specific mechanical signals may promote cell differentiation into a particular phenotype, potentially having an effect on embryonic development. The use of such mechanical signals in vitro in specially designed "bioreactors" may provide important adjuncts to standard biochemical signaling pathways for promoting engineered tissue growth. A further understanding of the biomechanical and biochemical pathways involved in mechanical signal transduction by stem cells will hopefully provide new insight for the improvement of stem-cell based therapies.
Collapse
Affiliation(s)
- Bradley T Estes
- Department of Surgery, Division of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
37
|
Mauney JR, Sjostorm S, Blumberg J, Horan R, O'Leary JP, Vunjak-Novakovic G, Volloch V, Kaplan DL. Mechanical stimulation promotes osteogenic differentiation of human bone marrow stromal cells on 3-D partially demineralized bone scaffolds in vitro. Calcif Tissue Int 2004; 74:458-68. [PMID: 14961210 DOI: 10.1007/s00223-003-0104-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2003] [Accepted: 10/03/2003] [Indexed: 12/13/2022]
Abstract
Bone is a dynamic tissue that is able to sense and adapt to mechanical stimuli by modulating its mass, geometry, and structure. Bone marrow stromal cells (BMSCs) are known to play an integral part in bone formation by providing an osteoprogenitor cell source capable of differentiating into mature osteoblasts in response to mechanical stresses. Characteristics of the in vivo bone environment including the three dimensional (3-D) lacunocanalicular structure and extracellular matrix composition have previously been shown to play major roles in influencing mechanotransduction processes within bone cells. To more accurately model this phenomenon in vitro, we cultured human BMSCs on 3-D, partially demineralized bone scaffolds in the presence of four-point bending loads within a novel bioreactor. The effect of mechanical loading and dexamethasone concentration on BMSC osteogenic differentiation and mineralized matrix production was studied for 8 and 16 days of culture. Mechanical stimulation after 16 days with 10 nM dexamethasone promoted osteogenic differentiation of BMSCs by significantly elevating alkaline phosphatase activity as well as alkaline phosphatase and osteopontin transcript levels over static controls. Mineralized matrix production also increased under these culture conditions. Dexamethasone concentration had a dramatic effect on the ability of mechanical stimulation to modulate these phenotypic and genotypic responses. These results provide increased insight into the role of mechanical stimulation on osteogenic differentiation of human BMSCs in vitro and may lead to improved strategies in bone tissue engineering.
Collapse
Affiliation(s)
- J R Mauney
- Tufts University, Department of Biomedical Engineering, Biotechnology Center, 4 Colby Street, Medford, Massachusetts 02155, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sakata T, Wang Y, Halloran BP, Elalieh HZ, Cao J, Bikle DD. Skeletal unloading induces resistance to insulin-like growth factor-I (IGF-I) by inhibiting activation of the IGF-I signaling pathways. J Bone Miner Res 2004; 19:436-46. [PMID: 15040832 PMCID: PMC10720400 DOI: 10.1359/jbmr.0301241] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2003] [Revised: 09/18/2003] [Accepted: 10/09/2003] [Indexed: 01/09/2023]
Abstract
UNLABELLED We showed that unloading markedly diminished the effects of IGF-I to activate its signaling pathways, and the disintegrin echistatin showed a similar block in osteoprogenitor cells. Furthermore, unloading decreased alphaVbeta3 integrin expression. These results show that skeletal unloading induces resistance to IGF-I by inhibiting activation of the IGF-I signaling pathways at least in part through downregulation of integrin signaling. INTRODUCTION We have previously reported that skeletal unloading induces resistance to insulin-like growth factor-I (IGF-I) with respect to bone formation. However, the underlying mechanism remains unclear. The aim of this study was to clarify how skeletal unloading induces resistance to the effects of IGF-I administration in vivo and in vitro with respect to bone formation. MATERIALS AND METHODS We first determined the response of bone to IGF-I administration in vivo during skeletal unloading. We then evaluated the response of osteoprogenitor cells isolated from unloaded bones to IGF-I treatment in vitro with respect to activation of the IGF-I signaling pathways. Finally we examined the potential role of integrins in mediating the responsiveness of osteoprogenitor cells to IGF-I. RESULTS IGF-I administration in vivo significantly increased proliferation of osteoblasts. Unloading markedly decreased proliferation and blocked the ability of IGF-I to increase proliferation. On a cellular level, IGF-I treatment in vitro stimulated the activation of its receptor, Ras, ERK1/2 (p44/42 MAPK), and Akt in cultured osteoprogenitor cells from normally loaded bones, but these effects were markedly diminished in cells from unloaded bones. These results were not caused by altered phosphatase activity or changes in receptor binding to IGF-I. Inhibition of the Ras/MAPK pathway was more impacted by unloading than that of Akt. The disintegrin echistatin (an antagonist of the alphaVbeta3 integrin) blocked the ability of IGF-I to stimulate its receptor phosphorylation and osteoblast proliferation, similar to that seen in cells from unloaded bone. Furthermore, unloading significantly decreased the mRNA levels both of alphaV and beta3 integrin subunits in osteoprogenitor cells. CONCLUSION These results indicate that skeletal unloading induces resistance to IGF-I by inhibiting the activation of IGF-I signaling pathways, at least in part, through downregulation of integrin signaling, resulting in decreased proliferation of osteoblasts and their precursors.
Collapse
Affiliation(s)
- Takeshi Sakata
- Department of Medicine, University of California, Endocrine Unit, Veterans Affairs Medical Center, San Francisco, California 94121, USA
| | | | | | | | | | | |
Collapse
|
39
|
Simmons CA, Matlis S, Thornton AJ, Chen S, Wang CY, Mooney DJ. Cyclic strain enhances matrix mineralization by adult human mesenchymal stem cells via the extracellular signal-regulated kinase (ERK1/2) signaling pathway. J Biomech 2003; 36:1087-96. [PMID: 12831733 DOI: 10.1016/s0021-9290(03)00110-6] [Citation(s) in RCA: 219] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Physical stimuli play critical roles in the development, regeneration, and pathology of many mesenchymal tissues, most notably bone. While mature bone cells, such as osteoblasts and osteocytes, are clearly involved in these processes, the role of their progenitors in mechanically mediated tissue responses is unknown. In this study, we investigated the effect of cyclic substrate deformation on the proliferation and osteogenic differentiation of human mesenchymal stem cells (hMSCs). Application of equibiaxial cyclic strain (3%, 0.25Hz) to hMSCs cultured in osteogenic media inhibited proliferation and stimulated a 2.3-fold increase in matrix mineralization over unstrained cells. The strain stimulus activated the extracellular signal-regulated kinase (ERK1/2) and p38 mitogen-activated protein kinase pathways, but had no effect on c-Jun N-terminal kinase phosphorylation or activity. Strain-induced mineralization was largely mediated by ERK1/2 signaling, as inhibition of ERK1/2 attenuated calcium deposition by 55%. Inhibition of the p38 pathway resulted in a more mature osteogenic phenotype, suggesting an inhibitory role for p38 signaling in the modulation of strain-induced osteogenic differentiation. These results demonstrate that mechanical signals regulate hMSC function, suggesting a critical role for physical stimulation of this specific cell population in mesenchymal tissue formation.
Collapse
Affiliation(s)
- Craig A Simmons
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
40
|
Sakata T, Halloran BP, Elalieh HZ, Munson SJ, Rudner L, Venton L, Ginzinger D, Rosen CJ, Bikle DD. Skeletal unloading induces resistance to insulin-like growth factor I on bone formation. Bone 2003; 32:669-80. [PMID: 12810174 DOI: 10.1016/s8756-3282(03)00088-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Skeletal unloading results in an inhibition of bone formation associated with a decrease in osteoblast number, impaired mineralization of bone, and altered proliferation and differentiation of osteoprogenitor cells. Although such changes are likely to be mediated by multiple factors, resistance to the growth-promoting action of insulin-like growth factor I (IGF-I) has been hypothesized to play an important role. To determine whether skeletal unloading induces resistance to IGF-I on bone formation, we examined the response of unloaded (hindlimb elevation) and normally loaded tibia and femur to IGF-I administration. To eliminate the variable of endogenous growth hormone production and secretion during exogenous IGF-I administration, we used growth hormone-deficient dwarf rats (dw-4). The rats were given IGF-I (2.5 mg/kg/day) or vehicle during 7 and 14 days of unloading or normal loading. This significantly increased the serum level of IGF-I in both the normally loaded and unloaded rats. Unloading did not affect the serum level of IGF-I in the vehicle-treated rats. IGF-I markedly increased periosteal bone formation at the tibiofibular junction of normally loaded rats. Unloading decreased bone formation in the vehicle-treated rats, and blocked the ability of IGF-I to increase bone formation. On the other hand, IGF-I increased periosteal bone formation at the midpoint of the humerus (normally loaded in this model) in both hindlimb-elevated and normally loaded rats. IGF-I significantly increased osteogenic colony number, total ALP activity, and total mineralization in bone marrow osteoprogenitor (BMOp) cells of normally loaded rats. Unloading reduced these parameters in the vehicle-treated rats, and blocked the stimulation by IGF-I. Furthermore, IGF-I administration (10 ng/ml) in vitro significantly increased cell proliferation of the BMOp cells isolated from normally loaded bone, but not that of cells from unloaded bone. These results indicate that skeletal unloading induces resistance to IGF-I on bone formation.
Collapse
Affiliation(s)
- T Sakata
- Department of Medicine, University of California, Veterans Affairs Medical Center, Endocrine Unit, San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Frank O, Heim M, Jakob M, Barbero A, Schäfer D, Bendik I, Dick W, Heberer M, Martin I. Real-time quantitative RT-PCR analysis of human bone marrow stromal cells during osteogenic differentiation in vitro. J Cell Biochem 2002; 85:737-46. [PMID: 11968014 DOI: 10.1002/jcb.10174] [Citation(s) in RCA: 281] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We developed and used real-time RT-PCR assays to investigate how the expression of typical osteoblast-related genes by human bone marrow stromal cells (BMSC) is regulated by (i) the culture time in medium inducing osteogenic differentiation and (ii) the previous expansion in medium enhancing cell osteogenic commitment. BMSC from six healthy donors were expanded in medium without (CTR) or with fibroblast growth factor-2 and dexamethasone (FGF/Dex; these factors are known to increase BMSC osteogenic commitment) and further cultivated for up to 20 days with ascorbic acid, beta-glycerophosphate and dexamethasone (these factors are typically used to induce BMSC osteogenic differentiation). Despite a high variability in the gene expression levels among different individuals, we identified the following statistically significant patterns. The mRNA levels of bone morphogenetic protein-2 (BMP-2), bone sialo protein-II (BSP), osteopontin (OP) and to a lower extent cbfa-1 increased with culture time in osteogenic medium (OM), both in CTR- and FGF/Dex-expanded BMSC, unlike levels of alkaline phosphatase, collagen type I, osteocalcin, and osteonectin. After 20 days culture in OM, BMP-2, BSP, and OP were more expressed in FGF/Dex than in CTR-expanded BMSC (mRNA levels were, respectively, 9.5-, 14.9-, and 5.8-fold higher), unlike all the other investigated genes. Analysis of single-colony-derived strains of BMSC further revealed that after 20 days culture in OM, only a subset of FGF/Dex-expanded clones expressed higher mRNA levels of BMP-2, BSP, and OP than CTR-expanded clones. In conclusion, we provide evidence that mRNA levels of BMP-2, BSP, and OP, quantified using real-time RT-PCR, can be used as markers to monitor the extent of BMSC osteogenic differentiation in vitro; using those markers, we further demonstrated that only a few subpopulations of BMSC display enhanced osteogenic differentiation following FGF/Dex expansion.
Collapse
Affiliation(s)
- Oliver Frank
- Department of Surgery, Research Division, University of Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shiiba M, Arnaud SB, Tanzawa H, Kitamura E, Yamauchi M. Regional alterations of type I collagen in rat tibia induced by skeletal unloading. J Bone Miner Res 2002; 17:1639-45. [PMID: 12211434 DOI: 10.1359/jbmr.2002.17.9.1639] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Skeletal unloading induces loss of mineral density in weight-bearing bones that leads to inferior bone mechanical strength. This appears to be caused by a failure of bone formation; however, its mechanisms still are not well understood. The objective of this study was to characterize collagen, the predominant matrix protein in bone, in various regions of tibia of rats that were subjected to skeletal unloading by 4 weeks tail suspension. Sixteen male Sprague-Dawley rats (4 months old) were divided into tail suspension and ambulatory controls (eight rats each). After the tail suspension, tibias from each animal were collected and divided into five regions and collagen was analyzed. The collagen cross-linking and the extent of lysine (Lys) hydroxylation in unloaded bones were significantly altered in proximal epiphysis, diaphysis, and, in particular, proximal metaphysis but not in distal regions. The pool of immature/nonmineralized collagen measured by its extractability with a chaotropic solvent was significantly increased in proximal metaphysis. These results suggest that skeletal unloading induced an accumulation of post-translationally altered nonmineralized collagen and that these changes are bone region specific. These alterations might be caused by impaired osteoblastic function/differentiation resulting in a mineralization defect.
Collapse
Affiliation(s)
- Masashi Shiiba
- Dental Research Center, University of North Carolina at Chapel Hill, 27599, USA
| | | | | | | | | |
Collapse
|
43
|
Watanuki M, Sakai A, Sakata T, Tsurukami H, Miwa M, Uchida Y, Watanabe K, Ikeda K, Nakamura T. Role of inducible nitric oxide synthase in skeletal adaptation to acute increases in mechanical loading. J Bone Miner Res 2002; 17:1015-25. [PMID: 12054156 DOI: 10.1359/jbmr.2002.17.6.1015] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To clarify the role of nitric oxide (NO) in regulation of bone metabolism in response to skeletal loading, we examined inducible NO synthase (iNOS) gene knockout mice in the tail-suspension model. Histomorphometric analyses of proximal tibias revealed that 7 days of tail suspension decreased the bone volume (BV/TV) and bone formation rate (BFR/BS) and increased the osteoclast surface (Oc.S/BS) in mice with all iNOS genotypes. Both iNOS+/+ and iNOS+/- mice responded to subsequent 14-day reloading, with increases in BV/TV and BFR/BS and a decrease in Oc.S/BS, whereas these responses were abolished in iNOS-/- mice. The osteoblasts flattened after tail suspension appeared cuboidal during subsequent reloading. Immunoreactivity for iNOS was detected in these osteoblasts and osteocytes by immunohistochemistry. These defective responses after reloading were rescued in iNOS-/- mice by treatment with an NO donor nitroglycerine (NG). Conversely, the responses in iNOS+/+ mice were inhibited by treatment with an NOS inhibitor aminoguanidine (AG). In bone marrow cell cultures, mineralized nodules derived from iNOS-/- mice after reloading were significantly reduced. Taken together, our results suggest that NO generated by iNOS in osteoblasts plays a critical role in adjusting bone turnover and increasing osteogenic activity in response to the acute increase in mechanical loading after tail suspension.
Collapse
Affiliation(s)
- Makoto Watanuki
- Department of Orthopedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ahdjoudj S, Lasmoles F, Holy X, Zerath E, Marie PJ. Transforming growth factor beta2 inhibits adipocyte differentiation induced by skeletal unloading in rat bone marrow stroma. J Bone Miner Res 2002; 17:668-77. [PMID: 11918224 DOI: 10.1359/jbmr.2002.17.4.668] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Skeletal unloading induced by hindlimb suspension in rats reduces bone formation and induces osteopenia, but its effect on adipogenesis is unknown. We assessed the effects of unloading and transforming growth factor (TGF) beta2 on bone marrow stromal cell adipocyte differentiation in relation with osteoblast differentiation. Skeletal unloading rapidly (4-7 days) decreased osteoblast transcription factor Runx2, osteocalcin (OC), and type I collagen messenger RNA (mRNA) levels and reduced bone formation in the long bone metaphysis. Conversely, unloading increased expression of the adipocyte transcription factor peroxisome proliferator-activated receptor gamma2 (PPARgamma2) at 4 days and increased expression of the adipocyte differentiation genes lipoprotein lipase (LPL) and aP2 in the bone marrow stroma at 7 days. Consistently, unloading increased the number and volume of adipocytes in the bone marrow stroma. Continuous (0-7 days) and late (4-7 days) treatments with TGF-beta2 corrected the abnormal expression of Cbfa1/Runx2, OC, and type I collagen mRNAs and normalized bone formation in unloaded metaphyseal bone. Moreover, both TGF-beta2 treatments decreased PPARy2 and C/EBPalpha mRNA levels at 4 days and normalized aP2 and LPL expression and adipocyte number and volume at 7 days. These results show that skeletal unloading increases adipocyte differentiation concomitantly with inhibition of osteoblast differentiation. These abnormalities are prevented and reversed by TGF-beta2, suggesting a role for TGF-beta in the control of adipogenic differentiation in the bone marrow stroma.
Collapse
Affiliation(s)
- Souhila Ahdjoudj
- Unit 349 INSERM, CNRS, Laboratory of Osteoblast Biology and Pathology, Lariboisière Hospital, Paris, France
| | | | | | | | | |
Collapse
|
45
|
Abstract
Normal loading strains of 200-2000 (mu)epsilon to bone result in bending forces, generating mechanical stretch and pressure gradients in canaliculi that drive extracellular fluid flow, resulting in stress on the membranes of osteocytes, lining cells, and osteoblasts. Under excess loading, as well as during unloading (e.g., microgravity, bed rest), the fluid shift and resultant change in interstitial fluid flow may play a larger role in bone remodeling than mechanical stretch. The in vitro model systems used to investigate mechanical loading of bone generate either fluid shear, hydrostatic compression, biaxial stretch, uniaxial stretch, or a combination of two or more of these forces. The results of in vitro experiments suggest that fluid shear is a major factor affecting bone cell metabolism. Both the flow-loop apparatus (which produces pulsatile flow and uses fluid shear as its principal stimulus) and the uniaxial silicone plate stretching apparatus (which generates cyclic stretch) create a reproducible and consistent stimulus. Endpoints measured in flow experiments, however, are short term and usually short lived, and it is unknown whether these changes impact the function of differentiated osteoblasts. Endpoints measured in uniaxial stretch experiments are generally long-term-sustained effects of mechanical perturbation and more easily relatable to changes in osteoblastic activity. Biaxial stretch devices create both bending and compressive forces, resulting in different types of force on the cells, with the relative amount of each depending on the position of the cell in the device. Therefore, systems that incorporate pulsatile fluid flow or uniaxial stretch as the principal stimulus should be further developed and implemented in the study of the relationship between mechanical loading and bone response.
Collapse
Affiliation(s)
- N Basso
- Faculty of Dentistry, University of Toronto, ON, Toronto, Canada.
| | | |
Collapse
|
46
|
Sakai A, Sakata T, Tanaka S, Okazaki R, Kunugita N, Norimura T, Nakamura T. Disruption of the p53 gene results in preserved trabecular bone mass and bone formation after mechanical unloading. J Bone Miner Res 2002; 17:119-27. [PMID: 11771658 DOI: 10.1359/jbmr.2002.17.1.119] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We tested the hypothesis that mechanical unloading facilitates signaling of p53, an important modulator of cell cycling and apoptosis, in bone marrow cells and thereby reduces trabecular bone volume (BV). We performed histomorphometric analyses and bone marrow cell cultures in tail-suspended (TS) p53 null (p53-/-) and wild-type (p53+/+) mice. Eight-week-old male mice were assigned to four groups after 1-week acclimatization: p53+/+ + ground control (GC), p53+/+ + TS, p53-/- + GC, and p53-/- + TS. Bilateral tibial samples were used for analysis. The histomorphometric parameters of trabecular structure, formation and resorption did not differ between the p53-/- + GC and p53+/+ + GC groups. Trabecular BV in p53+/+ + TS mice was significantly reduced to 45% of that in the p53+/+ + GC group after one week of TS. In contrast, BV in p53-/- + TS mice was preserved at the same level as that in the p53-/- + GC group. The bone formation rate (BFR) was significantly reduced in p53+/+ + TS but not in p53-/- + TS mice. Unloading significantly increased trabecular osteoclast number (Oc.N) and surface in p53+/+ + TS mice compared with the p53+/+ + GC group, but the difference was not significant between p53-/- + TS and p53-/- + GC mice. In bone marrow cell culture, the numbers of alkaline phosphatase-positive (ALP+) colony-forming units fibroblastic (CFU-f) and mineralized nodules were significantly reduced in p53+/+ + TS, but not p53-/- + TS mice. [3H]thymidine incorporation into bone marrow cells was higher in p53-/- mice than in p53+/+ mice, independent of mechanical loading or unloading. Flow cytometric cell cycle analysis revealed that unloading significantly increased the percentage of hypoploid bone marrow cells in p53+/+ mice relative to that in p53+/+ + GC mice, but there was no significant difference in ploidy between p53-/- + TS and p53-/- + GC mice. Expression levels of p53 and p21 mRNAs were enhanced after TS in bone marrow cells from p53+/+ mice. Our data show that trabecular bone mass and bone formation were preserved after tail-suspension in p53-/- mice, closely associated with ALP+ CFU-f and mineralized nodule formation in marrow cultures obtained from tibias of p53-/- mice. We speculate that bone loss due to mechanical unloading may be related to facilitation of intracellular p53-p21 signaling.
Collapse
Affiliation(s)
- Akinori Sakai
- Department of Orthopedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Vico L, Hinsenkamp M, Jones D, Marie PJ, Zallone A, Cancedda R. Osteobiology, strain, and microgravity. Part II: studies at the tissue level. Calcif Tissue Int 2001; 68:1-10. [PMID: 12037617 DOI: 10.1007/bf02684996] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/1999] [Accepted: 08/03/2000] [Indexed: 10/22/2022]
Abstract
Loading microgravity, and/or defective mechanical strain-forces have important effects on bone cells and bone quality and quantity. The complex mechanisms induced by strain and microgravity on bone cells have been reviewed in Part I of this paper. In Part II, we have considered the data on the alterations induced by unloading and microgravity on the skeleton and the mechanisms that are involved at the tissue level in animals and humans.
Collapse
Affiliation(s)
- L Vico
- INSERM E9901, LBBTO, Faculte de Médecine de St Etienne, France
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Electron microscopic techniques have been used to profile the morphologies of marrow sacs in different laboratory species. These structures all comprise a condensed layer of overlapping fibroblast-like stromal cells and apparently confine the medullary and endosteal osteoblast/lining cells to separate histiotypic compartments. There were some variations in the morphology of the sac cells in the different species. In rats, cats, and sheep, scanning electron microscopy (SEM) showed a seamless arrangement of marrow sac cells which resembled a thin, flat simple squamous epithelium; they displayed few intercellular cytoplasmic processes. In the rabbit and pigeon, the sac comprised a more woven, multilayered fabric of broadly elongate flat fibroblast-like cells which displayed numerous intercellular processes. Transmission electron microscopy (TEM) showed that all marrow sac cells were attenuated with elongated nuclei, a few small round mitochondria, and a sparse rough endoplasmic reticulum. In the majority of animals, the sac was one to two cell layers thick. The rabbit and pigeon sacs were multilayered, and never less than three to four cells deep. The cell layers were not closely apposed. Tight or gap junctions were absent at the points of intercellular contact. These morphological results suggest that marrow sacs are common elements of the vertebrate skeleton with species specific morphologies.
Collapse
Affiliation(s)
- L X Bi
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, Texas 77555-0892, USA.
| | | | | | | | | |
Collapse
|
49
|
Zerath E, Holy X, Roberts SG, Andre C, Renault S, Hott M, Marie PJ. Spaceflight inhibits bone formation independent of corticosteroid status in growing rats. J Bone Miner Res 2000; 15:1310-20. [PMID: 10893679 DOI: 10.1359/jbmr.2000.15.7.1310] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bone formation and structure have been shown repeatedly to be altered after spaceflight. However, it is not known whether these changes are related to a stress-related altered status of the corticosteroid axis. We investigated the role of corticosteroids on spaceflight-induced effects in rat pelvis and thoracic vertebrae. Thirty-six male Sprague-Dawley rats were assigned to a flight, flight control, or vivarium group (n = 12/group). Bilateral adrenalectomy was performed in six rats per group, the additional six rats undergoing sham surgery. Adrenalectomized (ADX) rats were implanted with corticosteroid pellets. On recovery from spaceflight, thoracic vertebrae and the whole pelvis were removed and processed for biochemistry, histomorphometry, or bone cell culture studies. The 17-day spaceflight resulted in decreased bone volume (BV) in the cotyle area of pelvic bones (-12%; p < 0.05) associated with approximately 50% inhibition of bone formation in the cancellous area of pelvic metaphyses and in thoracic vertebral bodies. The latter effect was associated with a decreased number of endosteal bone cells isolated from the bone surface (BS) in these samples (-42%; p < 0.05). This also was associated with a decreased number of alkaline phosphatase positive (ALP+) endosteal bone cells at 2 days and 4 days of culture, indicating decreased osteoblast precursor cell recruitment. Maintaining basal serum corticosterone levels in flight-ADX rats did not counteract the impaired bone formation in vertebral or pelvic bones. Moreover, the decreased ex vivo number of total and ALP+ endosteal bone cells induced by spaceflight occurred independent of endogenous corticosteroid hormone levels. These results indicate that the microgravity-induced inhibition of bone formation and resulting decreased trabecular bone mass in specific areas of weight-bearing skeleton in growing rats occur independently of endogenous glucocorticoid secretion.
Collapse
Affiliation(s)
- E Zerath
- INSERM Unité 349, Cell and Molecular Biology of Bone and Cartilage, Paris, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Bateman TA, Dunstan CR, Ferguson VL, Lacey DL, Ayers RA, Simske SJ. Osteoprotegerin mitigates tail suspension-induced osteopenia. Bone 2000; 26:443-9. [PMID: 10773583 DOI: 10.1016/s8756-3282(00)00256-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Osteoprotegerin (OPG) is a recently discovered protein related to the tumor necrosis factor receptor family. It has been shown to inhibit ovariectomy (ovx)-induced resorption in rats and increase bone mineral density in young mice. Tail suspension is a procedure that inhibits bone formation in maturing rodents. This study was designed to quantify OPG's effect on cortical bone formation. Fifty-four mice were assigned to one of five groups (n = 10-11/group). A baseline control group was killed on day 0 of the 10 day study. The remaining groups were: vivarium housed (nonsuspended) control mice receiving 0.3 mg/kg per day OPG; vivarium control mice receiving daily placebo injections; tail-suspended mice receiving 0. 3 mg/kg per day OPG; and tail-suspended mice receiving placebo injections. Tetracycline was administered on days 0 and 8. OPG treatment of tail-suspended mice produced mechanical properties similar to those of placebo-treated, vivarium-housed mice: structural stiffness (8.5%, 20.7%) and elastic (13.9%, 10.1%) and maximum (4.7%, 8.1%) force were increased compared with placebo controls (vivarium, suspended groups). Percent mineral composition was highly significantly greater (p < 0.001 for all comparisons) for OPG-treated mice in the femur, tibia, and humerus, relative to placebo treatment. Matrix mass was also significantly increased in the femur, although not to the same degree as mineral mass. OPG decreased the amount of femoral endocortical resorption compared with the placebo-treated groups for both vivarium (27%) and suspended (24%) mice. Administration of OPG significantly decreased endocortical formation of the tibia. Periosteal bone formation rates were not altered by OPG. OPG-mitigated tail suspension induced osteopenia not by returning bone formation to normal levels, but by inhibiting resorption and increasing percent mineral composition.
Collapse
Affiliation(s)
- T A Bateman
- BioServe Space Technologies, University of Colorado, Boulder, CO 80309-0429, USA.
| | | | | | | | | | | |
Collapse
|