1
|
Smit TH. Closing the osteon: Do osteocytes sense strain rate rather than fluid flow? Bioessays 2021; 43:e2000327. [PMID: 34111316 DOI: 10.1002/bies.202000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/05/2022]
Abstract
Osteons are cylindrical structures of bone created by matrix resorbing osteoclasts, followed by osteoblasts that deposit new bone. Osteons align with the principal loading direction and it is thought that the osteoclasts are directed by osteocytes, the mechanosensitive cells that reside inside the bone matrix. These osteocytes are presumably controlled by interstitial fluid flow, induced by the physiological loading of bones. Here I consider the stimulation of osteocytes while the osteon is closed by osteoblasts. In a conceptual finite element model, bone is considered a poro-elastic material and subjected to locomotion-induced loading conditions. It appears that the magnitude of flow is constant along the closing cone, while shear strain rate in the bone matrix diminishes linearly with the deposition of bone. This suggests that shear strain rate, rather than fluid flow, is the physical cue that controls osteocytes and bone deposition in newly formed osteons.
Collapse
Affiliation(s)
- Theodoor H Smit
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Department of Orthopaedic Surgery, Amsterdam University Medical Centers, Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Abstract
Bone is one of the most highly adaptive tissues in the body, possessing the capability to alter its morphology and function in response to stimuli in its surrounding environment. The ability of bone to sense and convert external mechanical stimuli into a biochemical response, which ultimately alters the phenotype and function of the cell, is described as mechanotransduction. This review aims to describe the fundamental physiology and biomechanisms that occur to induce osteogenic adaptation of a cell following application of a physical stimulus. Considerable developments have been made in recent years in our understanding of how cells orchestrate this complex interplay of processes, and have become the focus of research in osteogenesis. We will discuss current areas of preclinical and clinical research exploring the harnessing of mechanotransductive properties of cells and applying them therapeutically, both in the context of fracture healing and de novo bone formation in situations such as nonunion. Cite this article: Bone Joint Res 2019;9(1):1–14.
Collapse
|
3
|
Sheikh Z, Abdallah MN, Al-Jaf F, Chen G, Hamdan N, Young RN, Grynpas MD, Glogauer M. Improved bone regeneration using bone anabolic drug conjugates (C3 and C6) with deproteinized bovine bone mineral as a carrier in rat mandibular defects. J Periodontol 2020; 91:1521-1531. [PMID: 32100284 DOI: 10.1002/jper.19-0645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/03/2020] [Accepted: 02/06/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Deproteinized bovine bone mineral (DBBM) has been extensively studied and used for bone regeneration in oral and maxillofacial surgery. However, it lacks an osteoinductive ability. We developed two novel bone anabolic conjugated drugs, known as C3 and C6, of an inactive bisphosphonate and a bone activating synthetic prostaglandin agonist. The aim was to investigate whether these drugs prebound to DBBM granules have the potential to achieve rapid and enhanced bone regeneration. METHODS Bilateral defects (4.3 mm diameter circular through and through) were created in mandibular angles of 24 Sprague-Dawley rats were filled with DBBM Control, DBBM with C3 or DBBM with C6 (n = 8 defects per group/ each timepoint). After 2 and 4 weeks, postmortem samples were analyzed by microcomputed tomography followed by backscattering electron microscopy and histology. RESULTS DBBM grafts containing the C3 and C6 conjugated drugs showed significantly more bone formation than DBBM control at 2 and 4 weeks. The C6 containing DBBM demonstrated the highest percentage of new bone formation at 4 weeks. There was no significant difference in the percentage of the remaining graft between the different groups at 2 or 4 weeks. CONCLUSIONS DBBM granules containing conjugated drugs C3 and C6 induced greater new bone volume generated and increased the bone formation rate more than the DBBM controls. This is expected to allow the development of clinical treatments that provide more predictable and improved bone regeneration for bone defect repair in oral and maxillofacial surgery.
Collapse
Affiliation(s)
- Zeeshan Sheikh
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada.,Department of Dental Clinical Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mohamed-Nur Abdallah
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Faik Al-Jaf
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Gang Chen
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Nader Hamdan
- Department of Dental Clinical Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert N Young
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Marc D Grynpas
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Department of Dental Oncology and Maxillofacial Prosthetics, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Sheikh Z, Chen G, Al-Jaf F, Thévenin M, Banks K, Glogauer M, Young RN, Grynpas MD. In Vivo Bone Effects of a Novel Bisphosphonate-EP4a Conjugate Drug (C3) for Reversing Osteoporotic Bone Loss in an Ovariectomized Rat Model. JBMR Plus 2019; 3:e10237. [PMID: 31844825 PMCID: PMC6894726 DOI: 10.1002/jbm4.10237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/15/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023] Open
Abstract
Pathological bone loss is a regular feature of postmenopausal osteoporosis, and the microstructural changes along with the bone loss make the individual prone to getting hip, spine, and wrist fractures. We have developed a new conjugate drug named C3, which has a synthetic, stable EP4 agonist (EP4a) covalently linked to an inactive alendronate (ALN) that binds to bone and allows physiological remodeling. After losing bone for 12 weeks, seven groups of rats were treated for 8 weeks via tail‐vein injection. The groups were: C3 conjugate at low and high doses, vehicle‐treated ovariectomy (OVX) and sham, C1 (a similar conjugate, but with active ALN at high dose), inactive ALN alone, and a mixture of unconjugated ALN and EP4a to evaluate the conjugation effects. Bone turnover was determined by dynamic and static histomorphometry; μCT was employed to determine bone microarchitecture; and bone mechanical properties were evaluated via biomechanical testing. Treatment with C3 significantly increased trabecular bone volume and vertebral BMD versus OVX controls. There was also significant improvement in the vertebral load‐bearing abilities and stimulation of bone formation in femurs after C3 treatment. This preclinical research revealed that C3 resulted in significant anabolic effects on trabecular bone, and EP4a and ALN conjugation components are vital to conjugate anabolic efficacy. A combined therapy using an EP4 selective agonist anabolic agent linked to an inactive ALN is presented here that produces significant anabolic effects, allows bone remodeling, and has the potential for treating postmenopausal osteoporosis or other diseases where bone strengthening would be beneficial. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zeeshan Sheikh
- Lunenfeld-Tanenbaum Research Institute Mount Sinai Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathology University of Toronto Toronto Ontario Canada.,Faculty of Dentistry University of Toronto Toronto Ontario Canada.,Faculty of Dentistry Dalhousie University Halifax Nova Scotia Canada
| | - Gang Chen
- Department of Chemistry Simon Fraser University Burnaby British Columbia Canada
| | - Faik Al-Jaf
- Faculty of Dentistry University of Toronto Toronto Ontario Canada
| | - Marion Thévenin
- Department of Chemistry Simon Fraser University Burnaby British Columbia Canada
| | - Kate Banks
- Division of Comparative Medicine University of Toronto Toronto Ontario Canada.,Department of Physiology University of Toronto Toronto Ontario Canada
| | - Michael Glogauer
- Faculty of Dentistry University of Toronto Toronto Ontario Canada.,Department of Dental Oncology and Maxillofacial Prosthetics Princess Margaret Cancer Centre Toronto Ontario Canada
| | - Robert N Young
- Department of Chemistry Simon Fraser University Burnaby British Columbia Canada
| | - Marc D Grynpas
- Lunenfeld-Tanenbaum Research Institute Mount Sinai Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathology University of Toronto Toronto Ontario Canada.,Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
| |
Collapse
|
5
|
Foley KL, Hebela N, Keenan MA, Pignolo RJ. Histopathology of periarticular non-hereditary heterotopic ossification. Bone 2018; 109:65-70. [PMID: 29225159 DOI: 10.1016/j.bone.2017.12.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 11/26/2022]
Abstract
In the mature adult skeleton, new bone formation is normally restricted to regeneration of osseous tissue at sites of fracture. However, heterotopic ossification, or the formation of bone outside the normal skeleton, can occur within muscle, adipose, or fibrous connective tissue. Periarticular non-hereditary heterotopic ossification (NHHO) may occur after musculoskeletal trauma, following CNS injury, with certain arthropathies, or following injury or surgery that is often sustained in the context of age-related pathology. The histological mechanism of bone development in these forms of heterotopic ossification has thus far been uncharacterized. We performed a histological analysis of 90 bone specimens from 18 patients with NHHO secondary to defined precipitating conditions, including traumatic brain injury, spinal cord injury, cerebrovascular accident, trauma without neurologic injury, and total hip or knee arthroplasty. All bone specimens revealed normal endochondral osteogenesis at heterotopic sites. We defined the order of sequence progression in NHHO lesion formation as occurring through six distinct histological stages: (1) perivascular lymphocytic infiltration, (2) lymphocytic migration into soft tissue, (3) reactive fibroproliferation, (4) neovascularity, (5) cartilage formation, and (6) endochondral bone formation. This study provides the first systematic evaluation of the predominant histopathological findings associated with multiple forms of NHHO and shows that they share a common mechanism of lesion formation.
Collapse
Affiliation(s)
- Kristin L Foley
- Boston Osteopathic Health and University of Massachusetts Medical School, Worcester and Newton, MA, United States.
| | - Nader Hebela
- Neurological Institute, Cleveland Clinic Abu Dhabi, United Arab Emirates
| | - Mary Ann Keenan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
6
|
Kamal AF, Novriansyah R, Rahyussalim, Prabowo Y, Siregar NC. Fibrodysplasia Ossificans Progressiva: Difficulty in Diagnosis and Management A case report and literature review. J Orthop Case Rep 2016; 5:26-30. [PMID: 27299014 PMCID: PMC4719346 DOI: 10.13107/jocr.2250-0685.248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Introduction: Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant genetic disorder and characterized by postnatal progressive heterotopic ossification of the connective tissue. There are difficulties in diagnosing FOP, thus delayed or misdiagnosis and mismanagement is common. 3D printers have now become widely available and inexpensive, and can be used to rapidly produce life-size models based on CT scans of an individual patient. The availability of patient specific, accurate and detailed models of complex acetabular fractures can aid planning of surgical management on a patient specific basis. Case Report: We present the diagnosis and surgical management of a 9-year old Indonesian girl with FOP. She presented with extensive involvement of cervical spine and both shoulders. Total excision of occipito-cervico-lumbar and paravertebral ossification and also exostoses at bilateral shoulder was done. At three years follow up, she had local recurrence with similar range of movement of the shoulder and cervical spine. Conclusion: FOP is an extremely rare case. It is difficult to diagnose and manage FOP, therefore delayed or misdiagnosis and also inappropriate management is common.
Collapse
Affiliation(s)
- Achmad Fauzi Kamal
- Department of Orthopaedic and Traumatology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia
| | - Robin Novriansyah
- Department of Orthopaedic and Traumatology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia
| | - Rahyussalim
- Department of Orthopaedic and Traumatology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia
| | - Yogi Prabowo
- Department of Orthopaedic and Traumatology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia
| | - Nurjati Chairani Siregar
- Department of Anatomical Pathology, Cipto MAngunkusumo Hospital, Faculty of Medicine Universitas Indonesia
| |
Collapse
|
7
|
Hu S, Liu CC, Chen G, Willett T, Young RN, Grynpas MD. In vivo effects of two novel ALN-EP4a conjugate drugs on bone in the ovariectomized rat model for reversing postmenopausal bone loss. Osteoporos Int 2016; 27:797-808. [PMID: 26272313 DOI: 10.1007/s00198-015-3284-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/05/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Two alendronate-EP4 agonist (ALN-EP4a) conjugate drugs, C1 and C2, which differ in structure by a short linker molecule, were evaluated in ovariectomized (OVX) rats for their anabolic effects. We showed that C1 led to significant anabolic effects on cortical and trabecular bone while anabolic effects associated with C2 were minimal. INTRODUCTION EP4as were covalently linked to ALN to create ALN-EP4a conjugate anabolic bone drugs, C1 and C2, which differ in structure by a short linker molecule in C1. When administered systemically, C1 and C2 are delivered to bone through targeted binding of ALN, where local hydrolytic enzymes liberate EP4a from ALN to exert anabolic effects. Here, we compare effects of C1 to C2 in a curative in vivo study. METHODS Three-month-old female Sprague Dawley rats were OVX or sham operated and allowed to lose bone for 3 months. Animals were then treated via tail vein injections for 3 months and sacrificed. Treatment groups were as follows: C1L (5 mg/kg biweekly), C1H (5 mg/kg weekly), C2L (15 mg/kg monthly), C2H (15 mg/kg biweekly), OVX and sham control (phosphate-buffered saline (PBS) biweekly), and ALN/EP4a-unconjugated mixture (0.75 mg/kg each biweekly). RESULTS MicroCT analysis showed that C1H treatment significantly increased vertebral bone mineral density (vBMD) and trabecular bone volume versus OVX controls while C2 treatments did not. Biomechanical testing showed that C1H treatment but not C2 treatments led to significant improvement in the load bearing abilities of the vertebrae compared to OVX controls. C1 stimulated endocortical bone formation and increased load bearing in femurs, while C2 did not. CONCLUSIONS We showed that C1 led to significant anabolic effects on cortical and trabecular bone while anabolic effects associated with C2 were minimal. These results led us to hypothesize a mode of action by which presence of a linker is crucial in facilitating the anabolic effects of EP4a when dosed as a prodrug with ALN.
Collapse
MESH Headings
- Alendronate/therapeutic use
- Animals
- Bone Density/drug effects
- Bone Density Conservation Agents/therapeutic use
- Bone Remodeling/drug effects
- Disease Models, Animal
- Drug Combinations
- Drug Evaluation, Preclinical/methods
- Female
- Humans
- Lumbar Vertebrae/drug effects
- Lumbar Vertebrae/physiopathology
- Osteoporosis, Postmenopausal/drug therapy
- Osteoporosis, Postmenopausal/physiopathology
- Ovariectomy
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP4 Subtype/agonists
- Receptors, Prostaglandin E, EP4 Subtype/chemistry
- Receptors, Prostaglandin E, EP4 Subtype/therapeutic use
- Structure-Activity Relationship
- Weight-Bearing/physiology
- X-Ray Microtomography/methods
Collapse
Affiliation(s)
- S Hu
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - C C Liu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - G Chen
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - T Willett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - R N Young
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - M D Grynpas
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
- , 25 Orde St., Suite 417, Toronto, ON, M5T 3H7, Canada.
| |
Collapse
|
8
|
ZHAO QIANG, WANG XUEYAO, YU XIAOXIA, ZHAI YINGXIAN, HE XU, WU SHAN, SHI YINGAI. Expression of human telomerase reverse transcriptase mediates the senescence of mesenchymal stem cells through the PI3K/AKT signaling pathway. Int J Mol Med 2015; 36:857-64. [DOI: 10.3892/ijmm.2015.2284] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/08/2015] [Indexed: 11/06/2022] Open
|
9
|
Liu CC, Hu S, Chen G, Georgiou J, Arns S, Kumar NS, Young RN, Grynpas MD. Novel EP4 receptor agonist-bisphosphonate conjugate drug (C1) promotes bone formation and improves vertebral mechanical properties in the ovariectomized rat model of postmenopausal bone loss. J Bone Miner Res 2015; 30:670-80. [PMID: 25284325 DOI: 10.1002/jbmr.2382] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/29/2014] [Accepted: 09/11/2014] [Indexed: 11/12/2022]
Abstract
Current treatments for postmenopausal osteoporosis aim to either promote bone formation or inhibit bone resorption. The C1 conjugate drug represents a new treatment approach by chemically linking the antiresorptive compound alendronate (ALN) with the anabolic agent prostanoid EP4 receptor agonist (EP4a) through a linker molecule (LK) to form a conjugate compound. This enables the bone-targeting ability of ALN to deliver EP4a to bone sites and mitigate the systemic side effects of EP4a, while also facilitating dual antiresorptive and anabolic effects. In vivo hydrolysis is required to release the EP4a and ALN components for pharmacological activity. Our study investigated the in vivo efficacy of this drug in treating established bone loss using an ovariectomized (OVX) rat model of postmenopausal osteopenia. In a curative experiment, 3-month-old female Sprague-Dawley rats were OVX, allowed to lose bone for 7 weeks, then treated for 6 weeks. Treatment groups consisted of C1 conjugate at low and high doses, vehicle-treated OVX and sham, prostaglandin E2 (PGE2 ), and mixture of unconjugated ALN-LK and EP4a to assess the effect of conjugation. Results showed that weekly administration of C1 conjugate dose-dependently increased bone volume in trabecular bone, which partially or completely reversed OVX-induced bone loss in the lumbar vertebra and improved vertebral mechanical strength. The conjugate also dose-dependently stimulated endocortical woven bone formation and intracortical resorption in cortical bone, with high-dose treatment increasing the mechanical strength but compromising the material properties. Conjugation between the EP4a and ALN-LK components was crucial to the drug's anabolic efficacy. To our knowledge, the C1 conjugate represents the first time that a combined therapy using an anabolic agent and the antiresorptive compound ALN has shown significant anabolic effects which reversed established osteopenia.
Collapse
Affiliation(s)
- Careesa C Liu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Faghihi F, Papadimitropoulos A, Martin I, Eslaminejad MB. Effect of Purmorphamine on Osteogenic Differentiation of Human Mesenchymal Stem Cells in a Three-Dimensional Dynamic Culture System. Cell Mol Bioeng 2014. [DOI: 10.1007/s12195-014-0343-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
11
|
Liu X, Chen H, Zhu W, Chen H, Hu X, Jiang Z, Xu Y, Zhou Y, Wang K, Wang L, Chen P, Hu H, Wang C, Zhang N, Ma Q, Huang M, Hu D, Zhang L, Wu R, Wang Y, Xu Q, Yu H, Wang J. Transplantation of SIRT1-engineered aged mesenchymal stem cells improves cardiac function in a rat myocardial infarction model. J Heart Lung Transplant 2014; 33:1083-92. [PMID: 25034794 DOI: 10.1016/j.healun.2014.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 04/24/2014] [Accepted: 05/27/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previous studies have demonstrated that biological aging has a negative influence on the therapeutic effects of mesenchymal stem cells (MSCs)-based therapy. Using a rat myocardial infarction (MI) model, we tested the hypothesis that silent mating type information regulation 2 homolog 1 (SIRT1) may ameliorate the phenotype and improve the function of aged MSCs and thus enhance the efficacy of aged MSCs-based therapy. METHODS Sixty female rats underwent left anterior descending coronary artery ligation and were randomly assigned to receiving: intramyocardial injection of cell culture medium (DMEM group); SIRT1 overexpression vector-treated aged MSCs (SIRT1-aged MSCs group) obtained from aged male SD rats or empty vector-treated aged MSCs (vector-aged MSCs group). Another 20 sham-operated rats that underwent open-chest surgery without coronary ligation or any other intervention served as controls. RESULTS SIRT1-aged MSC group exhibited enhanced blood vessel density in the border zone of MI hearts, which was associated with reduced cardiac remodeling, leading to improved cardiac performance. Consistent with the in vivo data, our in vitro experiments also demonstrated that SIRT1 overexpression ameliorated aged MSCs senescent phenotype and recapitulated the pro-angiogenesis property of MSCs and conferred the anti-stress response capabilities, as indicated by increases in pro-angiogenic factors, angiopoietin 1 (Ang1) and basic fibroblast growth factor (bFGF), expressions and a decrease in anti-angiogenic factor thrombospondin-1 (TBS1) at mRNA levels, and increases in Bcl-2/Bax ratio at protein level. CONCLUSIONS Up-regulating SIRT1 expression could enhance the efficacy of aged MSCs-based therapy for MI as it relates to the amelioration of senescent phenotype and hence improved biological function of aged MSCs.
Collapse
Affiliation(s)
- Xianbao Liu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiqiang Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Jiang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinchuan Xu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhou
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kan Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihan Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Panpan Chen
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hengxun Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Na Zhang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qunchao Ma
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyuan Huang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dexing Hu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Zhang
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rongrong Wu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiyuan Xu
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Yu
- Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology, Key Division of Ministry of Health, Zhejiang University School of Medicine, Hangzhou, China; Provincial Key Laboratory of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
12
|
Chen H, Liu X, Zhu W, Chen H, Hu X, Jiang Z, Xu Y, Wang L, Zhou Y, Chen P, Zhang N, Hu D, Zhang L, Wang Y, Xu Q, Wu R, Yu H, Wang J. SIRT1 ameliorates age-related senescence of mesenchymal stem cells via modulating telomere shelterin. Front Aging Neurosci 2014; 6:103. [PMID: 24917814 PMCID: PMC4042159 DOI: 10.3389/fnagi.2014.00103] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/14/2014] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) senescence is an age-related process that impairs the capacity for tissue repair and compromises the clinical use of autologous MSCs for tissue regeneration. Here, we describe the effects of SIRT1, a NAD+-dependent deacetylase, on age-related MSCs senescence. Knockdown of SIRT1 in young MSCs induced cellular senescence and inhibited cell proliferation whereas overexpression of SIRT1 in aged MSCs reversed the senescence phenotype and stimulated cell proliferation. These results suggest that SIRT1 plays a key role in modulating age-induced MSCs senescence. Aging-related proteins, P16 and P21 may be downstream effectors of the SIRT1-mediated anti-aging effects. SIRT1 protected MSCs from age-related DNA damage, induced telomerase reverse transcriptase (TERT) expression and enhanced telomerase activity but did not affect telomere length. SIRT1 positively regulated the expression of tripeptidyl peptidase 1 (TPP1), a component of the shelterin pathway that protects chromosome ends from DNA damage. Together, the results demonstrate that SIRT1 quenches age-related MSCs senescence by mechanisms that include enhanced TPP1 expression, increased telomerase activity and reduced DNA damage.
Collapse
Affiliation(s)
- Huiqiang Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Xianbao Liu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Wei Zhu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Han Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Zhi Jiang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yinchuan Xu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Lihan Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yu Zhou
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Panpan Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Na Zhang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Dexing Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Ling Zhang
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yaping Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Qiyuan Xu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Rongrong Wu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Hong Yu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| |
Collapse
|
13
|
Faghihi F, Baghaban Eslaminejad M. The effect of nano-scale topography on osteogenic differentiation of mesenchymal stem cells. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:5-16. [DOI: 10.5507/bp.2013.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 02/14/2013] [Indexed: 01/08/2023] Open
|
14
|
Oh N, Kim S, Hosoya K, Okumura M. Compensatory cellular reactions to nonsteroidal anti-inflammatory drugs on osteogenic differentiation in canine bone marrow-derived mesenchymal stem cells. J Vet Med Sci 2014; 76:629-36. [PMID: 24419976 PMCID: PMC4073330 DOI: 10.1292/jvms.13-0482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The suppressive effects of nonsteroidal anti-inflammatory drugs (NSAIDs) on the
bone healing process have remained controversial, since no clinical data have clearly
shown the relationship between NSAIDs and bone healing. The aim of this study was to
assess the compensatory response of canine bone marrow-derived mesenchymal stem cells
(BMSCs) to several classes of NSAIDs, including carprofen, meloxicam, indomethacin and
robenacoxib, on osteogenic differentiation. Each of the NSAIDs (10 µM)
was administered during 20 days of the osteogenic process with human recombinant IL-1β (1
ng/ml) as an inflammatory stimulator. Gene expression
of osteoblast differentiation markers (alkaline phosphatase and osteocalcin), receptors of
PGE2 (EP2 and EP4) and enzymes for prostaglandin (PG) E2 synthesis (COX-1,
COX-2, cPGES and mPGES-1) was measured by using quantitative reverse
transcription-polymerase chain reaction. Protein production levels of alkaline
phosphatase, osteocalcin and PGE2 were quantified using an alkaline phosphatase
activity assay, osteocalcin immunoassay and PGE2 immunoassay, respectively.
Histologic analysis was performed using alkaline phosphatase staining, von Kossa staining
and alizarin red staining. Alkaline phosphatase and calcium deposition were suppressed by
all NSAIDs. However, osteocalcin production showed no significant suppression by NSAIDs.
Gene expression levels of PGE2-related receptors and enzymes were upregulated
during continuous treatment with NSAIDs, while certain channels for PGE2
synthesis were utilized differently depending on the kind of NSAIDs. These data suggest
that canine BMSCs have a compensatory mechanism to restore PGE2 synthesis,
which would be an intrinsic regulator to maintain differentiation of osteoblasts under
NSAID treatment.
Collapse
Affiliation(s)
- Namgil Oh
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | | | | | | |
Collapse
|
15
|
Faghihi F, Baghaban Eslaminejad M, Nekookar A, Najar M, Salekdeh GH. The effect of purmorphamine and sirolimus on osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. Biomed Pharmacother 2012; 67:31-8. [PMID: 23228449 DOI: 10.1016/j.biopha.2012.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 10/23/2012] [Indexed: 11/18/2022] Open
Abstract
Small molecules have been introduced as less expensive biologically active compounds that can regulate different developmental phenomena. Purmorphamine and sirolimus are two small molecules that, according to some studies, possess certain osteomodulatory effects. This study was set out to highlight the appropriate dose and response time of these small molecules on enhancement of osteogenesis in human bone marrow-derived mesenchymal stem cells from early to mid and late stages of differentiation. Alkaline phosphatase activity, matrix mineralization and expression of osteoblast genes were quantitatively assessed in vitro. For the in vivo study, we transplanted stem cell-based constructs subcutaneously into rats, and treated them daily with the most promising doses of the small molecule. The constructs were analyzed by real-time PCR and histological staining. Our results showed that Sirolimus reduced osteogenic differentiation of mesenchymal stem cells by decreasing alkaline phosphatase activity at dose of 100nM after 14 days and mineralization of the matrix at 14 and 21 days post-induction. Purmorphamine induced up-regulation of alkaline phosphatase activity and expression of RUNX-2 at day 14. Up-regulation of osteocalcin was detected at the 3 and 5μM doses of purmorphamine on day 14 post-induction. Matrix mineralization remained unchanged in the presence or absence of purmorphamine. This dose of small molecule also accelerated expression of Alkaline phosphatase transcripts in vivo. In conclusion, sirolimus had an inhibitory effect on osteogenic differentiation of human bone marrow-derived mesenchymal stem cells; while purmorphamine, particularly at a dose of 3μM, showed a promotive effect in vitro and in vivo.
Collapse
Affiliation(s)
- F Faghihi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | | | | | | |
Collapse
|
16
|
Stolzing A, Bauer E, Scutt A. Suspension cultures of bone-marrow-derived mesenchymal stem cells: effects of donor age and glucose level. Stem Cells Dev 2012; 21:2718-23. [PMID: 22462498 DOI: 10.1089/scd.2011.0406] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Both ageing and diabetes are associated with reduced numbers and functional viability of mesenchymal stem cells (MSCs) in vivo which in turn lead to degenerative pathologies of the musculoskeletal system. The overall aim of this study was to elucidate the effects of age and raised glucose levels on the proliferation and self-renewal of rat nonadherent bone marrow MSCs (Na-BM-MSCs) in suspension cultures. MSC cultures isolated from 3- and 12-month-old rats were maintained using the "pour-off" method for up to 14 days in media containing different glucose levels and the phenotype, growth characteristics, colony forming unit-fibroblastic (CFU-f) numbers, and pluripotency characteristics of these cells were determined. This study indicates that rat adult bone marrow harbors pluripotent Na-BM-MSCs that seem to be unaffected by ageing during in vitro expansion. The Na-BM-MSCs express the pluripotency markers Oct4, Sox2, and Nanog. It was found that culture in high-glucose-containing medium had a negative effect on colony formation and differentiation. In contrast to classical MSC cultures, the generation of colonies by Na-BM-MSCs in suspension culture was not reduced in the older animals. The Na-BM-MSCs were found to express the pluripotency markers Oct4, Sox2, and Nanog, suggesting a more primitive stage of differentiation as compared with adherent MSCs. These data indicate that rat adult bone marrow harbors a population of pluripotent Na-BM-MSCs that appear to be relatively unaffected by ageing during in vitro expansion in suspension.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Fraunhofer Institute for Cell Therapy & Immunology (IZI), Department of Cell Therapy, Leipzig, Germany.
| | | | | |
Collapse
|
17
|
Lee K, Kim H, Kim JM, Kim JR, Kim KJ, Kim YJ, Park SI, Jeong JH, Moon YM, Lim HS, Bae DW, Kwon J, Ko CY, Kim HS, Shin HI, Jeong D. Systemic transplantation of human adipose-derived stem cells stimulates bone repair by promoting osteoblast and osteoclast function. J Cell Mol Med 2012; 15:2082-94. [PMID: 21159123 PMCID: PMC4394219 DOI: 10.1111/j.1582-4934.2010.01230.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Systemic transplantation of adipose-derived stem cells (ASCs) is emerging as a novel therapeutic option for functional recovery of diverse damaged tissues. This study investigated the effects of systemic transplantation of human ASCs (hASCs) on bone repair. We found that hASCs secrete various bone cell-activating factors, including hepatocyte growth factor and extracellular matrix proteins. Systemic transplantation of hASCs into ovariectomized mice induced an increased number of both osteoblasts and osteoclasts in bone tissue and thereby prevented bone loss. We also observed that conditioned medium from hASCs is capable of stimulating proliferation and differentiation of osteoblasts via Smad/extracellular signal-regulated kinase (ERK)/JNK (c-jun NH2-terminal kinase) activation as well as survival and differentiation of osteoclasts via ERK/JNK/p38 activation in vitro. Overall, our findings suggest that paracrine factors secreted from hASCs improve bone repair and that hASCs can be a valuable tool for use in osteoporosis therapy.
Collapse
Affiliation(s)
- Kyunghee Lee
- Department of Microbiology, Yeungnam University College of Medicine, Daegu, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhang J, Wang JHC. BMP-2 mediates PGE(2) -induced reduction of proliferation and osteogenic differentiation of human tendon stem cells. J Orthop Res 2012; 30:47-52. [PMID: 21688312 PMCID: PMC3189415 DOI: 10.1002/jor.21485] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/27/2011] [Indexed: 02/04/2023]
Abstract
Tendon stem cells (TSCs) have been proposed to play a major role in the development of tendinopathy, which refers to pathological changes, such as calcification, in affected tendons. Using a human TSC (hTSC) culture model, this study investigated the effects of PGE(2) , an inflammatory mediator present in injured tendons, on hTSC proliferation and differentiation as well as the molecular mediator for such PGE(2) -induced effects. We found that PGE(2) treatment of hTSCs decreased cell proliferation and caused osteogenic differentiation of hTSCs in a dose-dependent manner. Also, PGE(2) treatment of hTSCs induced dose-dependent BMP-2 production in culture, and moreover, addition of BMP-2 to hTSC culture decreased cell proliferation and induced hTSC differentiation into osteoblasts. Finally, addition of BMP-2 antibodies to hTSC culture treated with PGE(2) nearly abolished PGE(2) effects on both cell proliferation and osteogenic differentiation. Taken together, the findings of this study showed that BMP-2 mediates PGE(2) -induced reduction of proliferation and osteogenic differentiation of hTSCs. We suggest that such a mechanism may be partially responsible for the formation of calcified tissues in tendinopathic tendons seen in clinical settings.
Collapse
Affiliation(s)
| | - James H-C. Wang
- Correspondence to: James H-C Wang, PhD, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 210 Lothrop Street, BST, E1640, Pittsburgh, PA 15213, Tel.: 412-648-9102; Fax: 412-648-8548,
| |
Collapse
|
19
|
Effect of age and diabetes on the response of mesenchymal progenitor cells to fibrin matrices. Int J Biomater 2011; 2011:378034. [PMID: 22194749 PMCID: PMC3238389 DOI: 10.1155/2011/378034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 08/23/2011] [Accepted: 09/04/2011] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cells are showing increasing promise in applications such as tissue engineering and cell therapy. MSC are low in number in bone marrow, and therefore in vitro expansion is often necessary. In vivo, stem cells often reside within a niche acting to protect the cells. These niches are composed of niche cells, stem cells, and extracellular matrix. When blood vessels are damaged, a fibrin clot forms as part of the wound healing response. The clot constitutes a form of stem cell niche as it appears to maintain the stem cell phenotype while supporting MSC proliferation and differentiation during healing. This is particularly appropriate as fibrin is increasingly being suggested as a scaffold meaning that fibrin-based tissue engineering may to some extent recapitulate wound healing. Here, we describe how fibrin modulates the clonogenic capacity of MSC derived from young/old human donors and normal/diabetic rats. Fibrin was prepared using different concentrations to modulate the stiffness of the substrate. MSC were expanded on these scaffolds and analysed. MSC showed an increased self-renewal on soft surfaces. Old and diabetic cells lost the ability to react to these signals and can no longer adapt to the changed environment.
Collapse
|
20
|
Way L, Scutt N, Scutt A. Cytocentrifugation: a convenient and efficient method for seeding tendon-derived cells into monolayer cultures or 3-D tissue engineering scaffolds. Cytotechnology 2011; 63:567-79. [PMID: 21948096 DOI: 10.1007/s10616-011-9391-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 08/12/2011] [Indexed: 02/06/2023] Open
Abstract
Tendon and ligament injuries are very common, requiring some 200,000 reconstructions per year in the USA. Autografting can be used to repair these but donor tissue is limited and harvesting leads to morbidity at the graft sites. Tissue engineering has been used to grow simple tissues such as skin, cartilage and bone and due to their low vascularity and simple structure, tendons should be ideal candidates for such an approach. Scaffolds are essential for tissue engineering as they provide structure and signals that regulate growth. However, they present a physical barrier to cell seeding with the majority of the cells congregating at the scaffold surface. To address this we used centrifugation to enhance penetration of tendon-derived cells to the centres of 3-D scaffolds. The process had no apparent deleterious effects on the cells and both plating efficiency and cell distribution improved. After attachment the cells continued to proliferate and deposit a collagenous matrix. Scaffold penetration was investigated using layers of Azowipes allowing the separation and examination of individual leaves. At relatively low g-forces, cells penetrated a stack of 6 Azowipes leaving cells attached to each leaf. These data suggest that cytocentrifugation improves the penetration and homogeneity of tendon derived cells in 3-D and monolayer cultures.
Collapse
Affiliation(s)
- Louise Way
- Bone Biology Group, Department of Human Metabolism, Faculty of Medicine, Dentistry and Health, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | | | | |
Collapse
|
21
|
Valenti MT, Giannini S, Donatelli L, Zanatta M, Bertoldo F, Sella S, Vilei MT, Ossi E, Realdi G, Lo Cascio V, Dalle Carbonare L. The effect of risedronate on osteogenic lineage is mediated by cyclooxygenase-2 gene upregulation. Arthritis Res Ther 2010; 12:R163. [PMID: 20738860 PMCID: PMC2945066 DOI: 10.1186/ar3122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 08/02/2010] [Accepted: 08/25/2010] [Indexed: 12/16/2022] Open
Abstract
Introduction The purpose of this study was to evaluate the effects of risedronate (Ris) in the modulation of bone formation in rats with glucocorticoid (GC)-induced osteoporosis by histomorphometric, immunohistochemical and gene expression analyses. Methods We analyzed structure, turnover and microarchitecture, cyclooxygenase 2 (COX-2) levels and osteocyte apoptosis in 40 female rats divided as follows: 1) vehicle of methylprednisolone (vGC) + vehicle of risedronate (vRis); 2) Ris 5 μg/Kg + vGC; 3) methylprednisolone (GC) 7 mg/Kg + vRis; 4) GC 7 mg/Kg +Ris 5 μg/Kg. In addition, we evaluated cell proliferation and expression of COX-2 and bone alkaline phosphatase (b-ALP) genes in bone marrow cells and MLO-y4 osteocytes treated with Ris alone or in co-treatment with the selective COX-2 inhibitor NS-398 or with dexametasone. Results Ris reduced apoptosis induced by GC of osteocytes (41% vs 86%, P < 0.0001) and increased COX-2 expression with respect to controls (Immuno-Hystochemical Score (IHS): 8.75 vs 1.00, P < 0.0001). These positive effects of Ris in bone formation were confirmed by in vitro data as the viability and expression of b-ALP gene in bone marrow cells resulted increased in a dose dependent manner. Conclusions These findings suggest a positive effect of Ris in bone formation and support the hypothesis that the up-regulation of COX-2 could be an additional mechanism of anabolic effect of Ris.
Collapse
|
22
|
Xu M, Choudhary S, Voznesensky O, Gao Q, Adams D, Diaz-Doran V, Wu Q, Goltzman D, Raisz LG, Pilbeam CC. Basal bone phenotype and increased anabolic responses to intermittent parathyroid hormone in healthy male COX-2 knockout mice. Bone 2010; 47:341-52. [PMID: 20471507 PMCID: PMC3056501 DOI: 10.1016/j.bone.2010.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 04/26/2010] [Accepted: 05/06/2010] [Indexed: 01/09/2023]
Abstract
Cyclooxygenase-2 (COX-2) knockout (KO) mice in inbred strains can have renal dysfunction with secondary hyperparathyroidism (HPTH), making direct effects of COX-2 KO on bone difficult to assess. COX-2 KO mice in an outbred CD-1 background did not have renal dysfunction but still had two-fold elevated PTH compared to wild type (WT) mice. Compared to WT mice, KO mice had increased serum markers of bone turnover, decreased femoral bone mineral density (BMD) and cortical bone thickness, but no differences in trabecular bone volume by microCT or dynamic histomorphometry. Because PTH is a potent inducer of COX-2 and prostaglandin (PG) production, we examined the effects of COX-2 KO on bone responses after 3 weeks of intermittent PTH. Intermittent PTH increased femoral BMD and cortical bone area more in KO mice than in WT mice and increased trabecular bone volume in the distal femur in both WT and KO mice. Although not statistically significant, PTH-stimulated increases in trabecular bone tended to be greater in KO mice than in WT mice. PTH increased serum markers of bone formation and resorption more in KO than in WT mice but increased the ratio of osteoblastic surface-to-osteoclastic surface only in KO mice. PTH also increased femoral mineral apposition rates and bone formation rates in KO mice more than in WT mice. Acute mRNA responses to PTH of genes that might mediate some anabolic and catabolic effects of PTH tended to be greater in KO than WT mice. We conclude that (1) the basal bone phenotype in male COX-2 KO mice might reflect HPTH, COX-2 deficiency or both, and (2) increased responses to intermittent PTH in COX-2 KO mice, despite the presence of chronic HPTH, suggest that absence of COX-2 increased sensitivity to PTH. It is possible that manipulation of endogenous PGs could have important clinical implications for anabolic therapy with PTH.
Collapse
Affiliation(s)
- Manshan Xu
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lee CM, Genetos DC, Wong A, Yellowley CE. Prostaglandin expression profile in hypoxic osteoblastic cells. J Bone Miner Metab 2010; 28:8-16. [PMID: 19471853 DOI: 10.1007/s00774-009-0096-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Accepted: 04/09/2009] [Indexed: 11/28/2022]
Abstract
Conditions such as fracture and unloading have been shown to be associated with tissue and cellular hypoxia in bone. The effects of hypoxia on bone cell physiology and ultimately its impact on bone tissue repair and remodeling are not well understood. In this study, we investigated the role of hypoxia on prostaglandin release from osteoblastic cells cultured in 2% (hypoxia), 5% (potentially cellular normoxia), and 21% (normoxia for standard cell culture conditions) oxygen for up to 24 h. We quantified the effects of reduced oxygen tension on the release of prostaglandin (PG)E(2), PGF(2alpha), PGD(2), and PGI(2). The mechanism by which hypoxia increases PG production was investigated by examining the various regulatory components of the PG biosynthetic pathway. Our data show that PGE(2) levels alone are significantly elevated under hypoxic conditions. Also, we show that cyclooxygenase (COX)-1 and COX-2 play an important role in hypoxia-induced PGE(2) production, possibly via a mechanism involving changes in their respective activity levels under low oxygen conditions. The effect of hypoxia on PGE(2) levels was mimicked by dimethyloxaloglycine, a known activator of the HIF pathway. In addition, we confirmed that HIF-1alpha was stabilized in osteoblastic cells under hypoxia. Taken together these data suggest a role for the HIF pathway in regulation of PGE(2) levels under hypoxic conditions. Previous studies have detected release of prostaglandins from areas of damaged bone, such as a fracture site, and our data may contribute to an understanding of how this release is regulated.
Collapse
Affiliation(s)
- Christina M Lee
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, 1321 Haring Hall, One Shields Ave, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
24
|
Kalyvas DG, Tarenidou M. Influence of nonsteroidal anti-inflammatory drugs on osseointegration. J Oral Sci 2009; 50:239-46. [PMID: 18818457 DOI: 10.2334/josnusd.50.239] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
This paper reviews contemporary literature concerning the possible influence of nonsteroidal anti-inflammatory drugs (NSAIDs) on osseointegration. In vitro studies concerning the effect of NSAIDs on growth factors and bone-generating cells are the primary source of data pertaining to this issue because relatively few in vivo studies have been conducted. It is concluded that prescribing NSAIDs during the early postoperative period is likely not without negative effect, although any negative influence appears to be temporary and does not affect the final outcome of osseointegration.
Collapse
Affiliation(s)
- Demos G Kalyvas
- Clinic of Oral and Maxillofacial Surgery, Dental School University of Athens, Athens, Greece.
| | | |
Collapse
|
25
|
Zhang ZL, Tong J, Lu RN, Scutt AM, Goltzman D, Miao DS. Therapeutic potential of non-adherent BM-derived mesenchymal stem cells in tissue regeneration. Bone Marrow Transplant 2009; 43:69-81. [PMID: 18711348 DOI: 10.1038/bmt.2008.260] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 07/07/2008] [Accepted: 07/18/2008] [Indexed: 12/16/2022]
Abstract
We demonstrated that non-adherent BM cells (NA-BMCs) can be expanded in suspension and give rise to multiple mesenchymal phenotypes including fibroblastic, osteoblastic, chondrocytic and adipocytic as well as glial cell lineages in vitro using the 'pour-off' BMC culture method. Mesenchymal stem cells (MSCs) derived from NA-BMCs (NA-MSCs) from wild-type mice were transplanted into VDR gene knockout (VDR(-/-)) mice that had received a lethal dose of radiation. Results revealed that NA-MSC can be used to rescue lethally irradiated mice and become incorporated into a diverse range of tissues. After lethal dose irradiation, all untransplanted mice died within 2 weeks, whereas those transplanted with NA-MSCs were viable for at least 3 months. Transplantation rescued these mice by reconstructing a hematopoietic system and repairing other damaged tissues. WBC, RBC and platelet counts recovered to normal after 1 month, and VDR gene expression was found in various tissues of viable VDR(-/-) recipients. Adult BM harbors pluripotent NA-MSCs, which can migrate in vivo into multiple body organs. In an appropriate microenvironment, they can adhere, proliferate and differentiate into specialized cells of target tissues and thus function in damaged tissue regeneration and repair.
Collapse
Affiliation(s)
- Z L Zhang
- Calcium Research Laboratory, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Sprowson AP, McCaskie AW, Birch MA. ASARM-truncated MEPE and AC-100 enhance osteogenesis by promoting osteoprogenitor adhesion. J Orthop Res 2008; 26:1256-62. [PMID: 18383145 DOI: 10.1002/jor.20606] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Matrix extracellular phosphoglycoprotein (MEPE) is a member of the SIBLING (Small Integrin-Binding Ligand, N-linked Glycoprotein) family of secreted glycophosphoproteins. Several previous studies have demonstrated that MEPE and its peptide motif, AC-100, may regulate bone mass and influence osteoblast activity, suggesting its potential for inclusion in novel therapeutic strategies aimed at increasing osteogenesis. Our study uses in vitro approaches to assess how adhesion of nonadherent cells is influenced by MEPE and whether response to MEPE is dependent on the maturity of osteoblastic cells. Truncated MEPE (ASARM removed) or AC-100 enhanced the adhesion, spreading, and focal complex formation of unadhered osteoblastic cells leading to increased differentiation and bone formation after 28 days of culture. Furthermore, addition of truncated MEPE or AC-100 to mature osteoblasts had no significant effect on bone formation. Our data supports an action for truncated MEPE and AC-100 in altering the physiology of immature poorly adherent cells which subsequently influences the way in which these cells interact with a substrate to facilitate their survival and/or commitment to the osteoblast lineage.
Collapse
Affiliation(s)
- Andrew P Sprowson
- Musculoskeletal Research Group, Institute for Cellular Medicine, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | |
Collapse
|
27
|
Weitao Y, Kangmei K, Xinjia W, Weili Q. Bone regeneration using an injectable calcium phosphate/autologous iliac crest bone composites for segmental ulnar defects in rabbits. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2008; 19:2485-2492. [PMID: 18253814 DOI: 10.1007/s10856-008-3383-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2007] [Accepted: 01/07/2008] [Indexed: 05/25/2023]
Abstract
BACKGROUND Treatment of segmental bone loss remains a challenge in skeletal repair. A major therapeutic goal is the development of implantable materials that will promote bone regeneration. OBJECTIVE We evaluate bone regeneration in grafts containing different concentrations autologous iliac crest bone (ACB) particles, carried in a new injectable calcium phosphate cement (CPC), in ulnar bone defects in rabbits. METHODS Large upper-mid-diaphyseal defects (10 mm) were created in the left ulnae of 60 skeletally mature New Zealand white rabbits. ACB concentrations of 0, 25, 50, 75, and 100% (by volume) in CPC were used to fill operated sites. Defect bridging was monitored by serial radiography at 4, 8, and 12 weeks post-operation. Samples were then examined histologically and by manual palpation to determine the extent of new bone formation. RESULTS At 4 weeks, we observed more elaborate structures and extensive absorption in ulnae treated with mixtures containing low concentrations of ACB (such as 0% and 25% volume of ACB/CPC), compared with those treated with mixtures containing high concentrations of ACB (such as 75% and 100% volume of ACB/CPC). At 8 weeks, histomorphometry revealed increased trabecular area and volume in the group treated with high ACB concentrations compared with those treated with low ACB concentrations. At 12 weeks, complete cortical bridging and regeneration of marrow space were detected in groups treated with high concentrations of ACB, and the amount of new bone regeneration was greater in these groups than in those treated with low ACB concentrations. CONCLUSIONS Treatment of rabbit ulnar defects with injectable CPC carrying an optimized concentration of ACB particles can lead to cortical bridging and bone marrow regeneration within 12 weeks.
Collapse
Affiliation(s)
- Yao Weitao
- Depertment of Orthopedics, He Nan Tumor Hospital, Dongming Road, Zhengzhou, He Nan Province 450000, China.
| | | | | | | |
Collapse
|
28
|
Abstract
Osteoblasts are the cells responsible for formation of new bone throughout life. Rats are one of the most widely studied mammalian species in skeletal biology and serve as useful models for many aspects of human skeletal physiology. The availability of genetically modified mice as research tools has greatly enabled our understanding of how specific genes contribute to the process of skeletogenesis. In order to explore the impact of biochemical, genetic, or pharmacological manipulation on bone formation, various osteogenic cell culture systems have been developed. Two of the most widely accepted rodent osteogenic culture models, using osteoprogenitor cells isolated from calvaria or bone marrow, are described in this chapter.
Collapse
Affiliation(s)
- Thomas A Owen
- Department of Cardiovascular, Metabolic and Endocrine Diseases, Pfizer Global Research and Development, Groton, CT, USA
| | | |
Collapse
|
29
|
Knippenberg M, Helder MN, de Blieck-Hogervorst JMA, Wuisman PIJM, Klein-Nulend J. Prostaglandins differentially affect osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. ACTA ACUST UNITED AC 2007; 13:2495-503. [PMID: 17655490 DOI: 10.1089/ten.2006.0420] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are currently used for bone tissue engineering. AT-MSCs undergoing osteogenic differentiation respond to mechanical loading with increased cyclooxygenase-2 gene expression, a key enzyme in prostaglandin (PG) synthesis. PGs are potent multifunctional regulators in bone, exhibiting stimulatory and inhibitory effects on bone formation and resorption. PGE(2), but not PGI(2) or PGF(2), recruits osteoprogenitors from the bone marrow space and influences their differentiation. We hypothesize that PGE(2), PGI(2), and PGF(2) may differentially regulate osteogenic differentiation of human AT-MSCs. PGE(2), PGI(2), and PGF(2) (0.01-10 microM) affected osteogenic differentiation, but not proliferation of AT-MSCs after 4-14 days. Only PGF(2) (0.01-10 microM) increased alkaline phosphatase (ALP) activity at day 4. PGE(2) (10 microM), PGI(2) (0.01-10 microM), and PGF(2) (10 microM) decreased ALP activity, whereas PGF(2) (0.1 microM) increased ALP activity at day 14. PGF(2) (0.01-0.1 microM) and PGI(2) (0.01 microM) upregulated osteopontin gene expression, and PGF(2) (0.01 microM) upregulated alpha1(I)procollagen gene expression at day 4. PGE(2) and PGF(2) (10 microM) at day 4 and PGF(2) (1 microM) at day 14 downregulated runt-related transcription factor-2 gene expression. We conclude that PGE(2), PGI(2), and PGF(2) differentially affect osteogenic differentiation of AT-MSCs, with PGF(2) being the most potent. Thus, locally produced PGF(2) might be most beneficial in promoting osteogenic differentiation of AT-MSCs, resulting in enhanced bone formation for bone tissue engineering.
Collapse
Affiliation(s)
- M Knippenberg
- Department of Oral Cell Biology, Academic Center of Dentistry Amsterdam (ACTA)-Universiteit van Amsterdam and Vrije Universiteit, Research Institute MOVE, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Stringer B, Waddington R, Sloan A, Phillips I, Telford G, Hughes D, Craig G, Gangemi L, Brook I, Freeman C, Cao X, Gosal M, Smith S, Russell G, Foster G. Bespoke human hypertrophic chondrocytic cell lines provide the osteoinductive signals required for vascularized bone formation. ACTA ACUST UNITED AC 2007; 13:133-45. [PMID: 17518587 DOI: 10.1089/ten.2006.0111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hypertrophic cartilage provides the morphological and biochemical template for orchestrating bone growth. To produce a bone-inductive material such as hypertrophic cartilage for clinical use, we have conditionally immortalized hypertrophic chondrocytic cells from human femur and expanded them in vitro through more than 145 divisions. The clonal cell lines generated by this process consistently express signals that induce both rat and human marrow cells to differentiate in vitro into osteoblastic cells. Further, implantation of the cell-free extracellular matrix from the immortalized chondrocytic cells causes vascularized bone to form in vivo in bony defects, but not in ectopic sites such as skeletal muscle. This study shows that molecular techniques can be used to generate bespoke human cell lines for bone tissue engineering. It also demonstrates that matrix material generated from human immortalized hypertrophic chondrocytic cells may provide an abundant, efficacious, and safer alternative to bone autograft--the currently preferred material for fracture repair.
Collapse
|
31
|
Xu Z, Choudhary S, Okada Y, Voznesensky O, Alander C, Raisz L, Pilbeam C. Cyclooxygenase-2 gene disruption promotes proliferation of murine calvarial osteoblasts in vitro. Bone 2007; 41:68-76. [PMID: 17467356 PMCID: PMC1993538 DOI: 10.1016/j.bone.2007.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Revised: 02/21/2007] [Accepted: 03/12/2007] [Indexed: 12/31/2022]
Abstract
Cyclooxygenase-2 (COX-2) is highly expressed in osteoblasts, and COX-2 produced prostaglandins (PGs) can increase osteoblastic differentiation in vitro. The goal of this study was to examine effects of COX-2 expression on calvarial osteoblastic proliferation and apoptosis. Primary osteoblasts (POBs) were cultured from calvariae of COX-2 wild-type (WT) and knockout (KO) mice. POB proliferation was evaluated by (3)H-thymidine incorporation and analysis of cell replication and cell cycle distribution by flow cytometry. POB apoptosis was evaluated by annexin and PI staining on flow cytometry. As expected, PGE(2) production and alkaline phosphatase (ALP) activity were increased in WT cultures compared to KO cultures. In contrast, cell numbers were decreased in WT compared to KO cells by day 4 of culture. Proliferation, measured on days 3-7 of culture, was 2-fold greater in KO than in WT POBs and associated with decreased Go/G1 and increased S cell cycle distribution. There was no significant effect of COX-2 genotype on apoptosis under basal culture conditions on day 5 of culture. Cell growth was decreased in KO POBs by the addition of PGE(2) or a protein kinase A agonist and increased in WT POBs by the addition of NS398, a selective COX-2 inhibitor. In contrast, differentiation and cell growth in marrow stromal cell (MSC) cultures, evaluated by ALP and crystal violet staining respectively, were increased in MSCs from WT mice compared to MSCs from KO mice, and exogenous PGE(2) increased cell growth in KO MSC cultures. We conclude that PGs secondary to COX-2 expression decrease osteoblastic proliferation in cultured calvarial cells but increase growth of osteoblastic precursors in MSC cultures.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Shilpa Choudhary
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Yosuke Okada
- First department of Internal Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555 Japan
| | - Olga Voznesensky
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Cynthia Alander
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Lawrence Raisz
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Carol Pilbeam
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
32
|
Choudhary S, Halbout P, Alander C, Raisz L, Pilbeam C. Strontium ranelate promotes osteoblastic differentiation and mineralization of murine bone marrow stromal cells: involvement of prostaglandins. J Bone Miner Res 2007; 22:1002-10. [PMID: 17371157 DOI: 10.1359/jbmr.070321] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Strontium ranelate is a new anti-osteoporosis treatment. This study showed that strontium ranelate stimulated PGE(2) production and osteoblastic differentiation in murine marrow stromal cells, which was markedly reduced by inhibition of COX-2 activity or disruption of COX-2 gene expression. Hence, some anabolic effects of strontium ranelate may be mediated by the induction of COX-2 and PGE(2) production. INTRODUCTION Strontium ranelate is an orally active drug that reduces vertebral and hip fracture risk by increasing bone formation and reducing bone resorption. Strontium ranelate effects on bone formation are the result of increased osteoblastic differentiation and activity, but the mechanisms governing these effects are unknown. Based on previous work, we hypothesized that strontium ranelate increases cyclooxygenase (COX)-2 expression and that, consequently, the prostaglandin E(2) (PGE(2)) produced could mediate some effects of strontium ranelate on osteoblasts. MATERIALS AND METHODS Marrow stromal cells (MSCs) from COX-2 wildtype (WT) and knockout (KO) mice were cultured with and without low-dose dexamethasone. Osteoblastic differentiation was characterized by alkaline phosphatase (ALP) activity, real-time PCR for ALP and osteocalcin (OCN) mRNA expression, and alizarin red staining for mineralization. Medium PGE(2) was measured by radioimmunoassay or enzyme immunoassay. RESULTS AND CONCLUSIONS In MSCs from COX-2 WT mice, strontium ranelate significantly increased ALP activity, ALP and OCN mRNA expression, and mineralization after 14 or 21 days of culture. A short treatment at the beginning of the culture (0-7 days) with strontium ranelate was as effective as continuous treatment. Strontium ranelate (1 and 3 mM Sr(+2)) dose-dependently increased PGE(2) production, with maximum PGE(2) production occurring during the first week of culture. NS-398, a selective COX-2 inhibitor, blocked the strontium ranelate stimulation of PGE(2) production and significantly inhibited the strontium ranelate stimulation of ALP activity. In MSCs from COX-2 KO mice, the strontium ranelate stimulation of ALP and OCN mRNA expression and mineralization were markedly reduced compared with COX-2 WT cultures. Similar effects of strontium ranelate on osteoblastic markers and on PGE(2) production were seen when MSCs were cultured with or without low-dose dexamethasone (10 nM). We conclude that PGE(2) produced by the strontium ranelate induction of COX-2 expression plays a role in strontium ranelate-induced osteoblastic differentiation in MSCs in vitro.
Collapse
Affiliation(s)
- Shilpa Choudhary
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | | | | | |
Collapse
|
33
|
Marie PJ, Fromigué O. Osteogenic differentiation of human marrow-derived mesenchymal stem cells. Regen Med 2007; 1:539-48. [PMID: 17465848 DOI: 10.2217/17460751.1.4.539] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are adherent cells that differentiate into chondroblasts, osteoblasts and adipocytes. In this short review, we summarize the molecular mechanisms that are known to control osteoblast differentiation and osteogenic potential of MSCs in vitro. We discuss the advances made in gene-based therapy to promote osteogenic differentiation of MSCs and the perspectives for an optimal use of MSCs for bone tissue regeneration or repair. One important challenge at the present time is to identify factors and pathways that promote osteogenic commitment of MSCs in order to use MSCs with functional potential for optimal bone repair in humans. In this context, genomic and proteomic analyses may help to identify molecules that could be used to promote osteogenic differentiation of human MSCs. In the future this may lead to selective therapeutic strategies for tissue engineering application in bone regeneration and repair in humans.
Collapse
Affiliation(s)
- Pierre J Marie
- Laboratory of Osteoblast Biology and Pathology Unité 606 INSERM, Hopital Lariboisière, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France.
| | | |
Collapse
|
34
|
Oreffo ROC, Cooper C, Mason C, Clements M. Mesenchymal stem cells: lineage, plasticity, and skeletal therapeutic potential. ACTA ACUST UNITED AC 2007; 1:169-78. [PMID: 17142852 DOI: 10.1385/scr:1:2:169] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tremendous capacity of bone to regenerate is indicative of the presence of stem cells with the capability, by definition, to self-renew as well as to give rise to daughter cells. These primitive progenitors, termed mesenchymal stem cells or bone marrow stromal stem cells, exist postnatally, and are multipotent with the ability to generate cartilage, bone, muscle, tendon, ligament, and fat. Given the demographic challenge of an ageing population, the development of strategies to exploit the potential of stem cells to augment bone formation to replace or restore the function of traumatized, diseased, or degenerated bone is a major clinical and socioeconomic need. Owing to the developmental plasticity of mesenchymal stem cells, there is great interest in their application to replace damaged tissues. Combined with modern advances in gene therapy and tissue engineering, they have the potential to improve the quality of life for many. Critical in the development of this field will be an understanding of the phenotype, plasticity, and potentiality of these cells and the tempering of patients' expectations driven by commercial and media hype to match current laboratory and clinical observations.
Collapse
Affiliation(s)
- Richard O C Oreffo
- Bone and Joint Research Group, University of Southampton, Southampton S016 6YD, UK.
| | | | | | | |
Collapse
|
35
|
Scutt A, Williamson EM. Cannabinoids stimulate fibroblastic colony formation by bone marrow cells indirectly via CB2 receptors. Calcif Tissue Int 2007; 80:50-9. [PMID: 17205329 DOI: 10.1007/s00223-006-0171-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 09/23/2006] [Indexed: 12/23/2022]
Abstract
Recently, the cannabinoid receptors CB(1) and CB(2) were shown to modulate bone formation and resorption in vivo, although little is known of the mechanisms underlying this. The effects of cannabinoids on mesenchymal stem cell (MSC) recruitment in whole bone marrow were investigated using either the fibroblastic colony-forming unit (CFU-f) assay or high-density cultures of whole bone marrow. Levels of the CB(1) and CB(2) receptors were assessed by flow cytometry. Treatment of CFU-f cultures with the endocannabinoid 2-arachidonylglycerol (2-AG) dose-dependently increased fibroblastic and differentiated colony formation along with colony size. The nonspecific agonists CP 55,940 and WIN 55,212 both increased colony numbers, as did the CB(2) agonists BML190 and JWH015. The CB(1)-specific agonist ACEA had no effect, whereas the CB(2) antagonist AM630 blocked the effect of the natural cannabinoid tetrahydrocannabivarin, confirming mediation via the CB(2) receptor. Treatment of primary bone marrow cultures with 2-AG stimulated proliferation and collagen accumulation, whereas treatment of subcultures of MSC had no effect, suggesting that the target cell is not the MSC but an accessory cell present in bone marrow. Subcultures of MSCs were negative for CB(1) and CB(2) receptors as shown by flow cytometry, whereas whole bone marrow contained a small population of cells positive for both receptors. These data suggest that cannabinoids may stimulate the recruitment of MSCs from the bone marrow indirectly via an accessory cell and mediated via the CB(2) receptor. This recruitment may be one mechanism responsible for the increased bone formation seen after cannabinoid treatment in vivo.
Collapse
MESH Headings
- Animals
- Arachidonic Acids/pharmacology
- Benzoxazines
- Bone Marrow Cells/cytology
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/physiology
- Cannabinoid Receptor Modulators/pharmacology
- Cannabinoids/pharmacology
- Cells, Cultured
- Colony-Forming Units Assay
- Cyclohexanols/pharmacology
- Dose-Response Relationship, Drug
- Endocannabinoids
- Glycerides/pharmacology
- Indomethacin/analogs & derivatives
- Indomethacin/pharmacology
- Male
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/physiology
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/physiology
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/drug effects
- Receptor, Cannabinoid, CB2/physiology
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/physiology
Collapse
Affiliation(s)
- A Scutt
- Tissue Engineering Group, Department of Engineering Materials, Kroto Research Institute Nanoscience and Technology Centre, University of Sheffield, Broad Lane, Sheffield S3 7HQ, UK.
| | | |
Collapse
|
36
|
Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res 2006; 327:449-62. [PMID: 17053900 DOI: 10.1007/s00441-006-0308-z] [Citation(s) in RCA: 505] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2006] [Accepted: 07/17/2006] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are increasingly being reported as occurring in a variety of tissues. Although MSCs from human bone marrow are relatively easy to harvest, the isolation of rodent MSCs is more difficult, thereby limiting the number of experiments in vivo. To determine a suitable cell source, we isolated rat MSCs from bone marrow, synovium, periosteum, adipose, and muscle and compared their properties for yield, expansion, and multipotentiality. After two passages, the cells in each population were CD11b (-), CD45 (-), and CD90 (+). The colony number per nucleated cells derived from synovium was 100-fold higher than that for cells derived from bone marrow. With regard to expansion potential, synovium-derived cells were the highest in colony-forming efficiency, fold increase, and growth kinetics. An in vitro chondrogenesis assay demonstrated that the pellets derived from synovium were heavier, because of their greater production of cartilage matrix, than those from other tissues, indicating their superiority in chondrogenesis. Synovium-derived cells retained their chondrogenic potential after a few passages. The Oil Red-O positive colony-rate assay demonstrated higher adipogenic potential in synovium- and adipose-derived cells. Alkaline phosphatase activity was greater in periosteum- and muscle-derived cells during calcification. The yield and proliferation potential of rat MSCs from solid tissues was much better than those from bone marrow. In particular, synovium-derived cells had the greatest potential for both proliferation and chondrogenesis, indicating their usefulness for cartilage study in a rat model.
Collapse
Affiliation(s)
- Hideya Yoshimura
- Section of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Weinreb M, Shamir D, Machwate M, Rodan GA, Harada S, Keila S. Prostaglandin E2 (PGE2) increases the number of rat bone marrow osteogenic stromal cells (BMSC) via binding the EP4 receptor, activating sphingosine kinase and inhibiting caspase activity. Prostaglandins Leukot Essent Fatty Acids 2006; 75:81-90. [PMID: 16890416 DOI: 10.1016/j.plefa.2006.06.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 06/05/2006] [Accepted: 06/08/2006] [Indexed: 11/23/2022]
Abstract
Prostaglandin E(2) (PGE(2)) is bone-anabolic, i.e. stimulates bone formation and increases bone mass. In this study, we explored possible intracellular mechanisms of its increase of osteogenic cells in rat bone marrow. Adherent rat bone marrow cells were counted after 12-48 h or cultured for 21 days and mineralized nodules were counted. Also, apoptosis of marrow cells was measured after in vivo PGE(2) injection. PGE(2) (100 nM) increased 2-3 fold the number of adherent BMSC, an effect which was mediated via binding the EP(4) receptor since it was mimicked by forskolin and 11-deoxy-prostaglandin E(1) (PGE(1)) and was blocked by DDA and L-161982 (EP(4) antagonist). PGE(2) stimulated sphingosine kinase (SPK) activity since its effects were blocked by DMS (SPK inhibitor) and mimicked by SPP (SPK product). PGE(2) reduced the activity of caspase-3 and -8 in BMSC and their inhibitors increased BMSC number and nodule formation. In vivo, PGE(2) prevented the increase in the apoptosis of bone marrow cells caused by indomethacin. We propose that PGE(2) exerts an anti-apoptotic effect on BMSC, thereby increasing their number and subsequent osteoblastic differentiation. Such an effect could explain how PGE(2) stimulates bone formation in vivo.
Collapse
Affiliation(s)
- M Weinreb
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.
| | | | | | | | | | | |
Collapse
|
38
|
Stolzing A, Scutt A. Effect of reduced culture temperature on antioxidant defences of mesenchymal stem cells. Free Radic Biol Med 2006; 41:326-38. [PMID: 16814114 DOI: 10.1016/j.freeradbiomed.2006.04.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 04/06/2006] [Accepted: 04/14/2006] [Indexed: 11/18/2022]
Abstract
Mesenchymal stem cells (MSC) promise to be valuable therapeutic tools but, due to their low numbers, require considerable in vitro expansion before use. This leads to in vitro aging, the accumulation of intracellular oxidative damage, and subsequently a decreased potential for proliferation and differentiation. Optimised culture conditions might help to reduce oxidative damage in MSC in vitro, and therefore, as reduced temperature is known to reduce oxidative stress in other somatic cells, we have investigated the effect of reduced temperature on rat MSC viability, differentiation, and oxidative damage. Temperature reduction did not affect MSC viability but increased differentiation and reduced apoptosis. Oxidative-damage-related indices were improved; reactive oxide species, nitric oxide, thiobarbituric acid reactive substances, carbonyl, and lipofuscin levels were reduced and glutathione peroxidase and superoxide dimutase levels increased. Levels of antiapoptotic heat shock proteins (HSP-27, -70, and -90) were raised and levels of the proapoptotic HSP-60 reduced. These data demonstrate that culturing MSC at reduced temperature decreases the accumulation of oxidative damage and therefore would probably improve long-term viability and successful engraftment of MSC used for tissue engineering or cell therapeutic purposes.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Kroto Research Institute, Department of Engineering Materials, University of Sheffield, Sheffield S3 7HQ, UK.
| | | |
Collapse
|
39
|
Abstract
In most mesenchymal tissues a subcompartment of multipotent progenitor cells is responsible for the maintenance and repair of the tissue following trauma. With increasing age, the ability of tissues to repair themselves is diminished, which may be due to reduced functional capacity of the progenitor cells. The purpose of this study was to investigate the effect of aging on rat mesenchymal progenitor cells. Mesenchymal progenitor cells were isolated from Wistar rats aged 3, 7, 12 and 56 weeks. Viability, capacity for differentiation and cellular aging were examined. Cells from the oldest group accumulated raised levels of oxidized proteins and lipids and showed decreased levels of antioxidative enzyme activity. This was reflected in decreased fibroblast colony-forming unit (CFU-f) numbers, increased levels of apoptosis and reduced proliferation and potential for differentiation. These data suggest that the reduced ability to maintain mesenchymal tissue homeostasis in aged mammals is not purely due to a decline in progenitor cells numbers but also to a loss of progenitor functionality due to the accumulation of oxidative damage, which may in turn be a causative factor in a number of age-related pathologies such as arthritis, tendinosis and osteoporosis.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Centre for Biomaterials and Tissue Engineering, Department of Engineering Materials, University of Sheffield, North Campus, Sheffield, S3 7HQ, UK.
| | | |
Collapse
|
40
|
Stolzing A, Coleman N, Scutt A. Glucose-induced replicative senescence in mesenchymal stem cells. Rejuvenation Res 2006; 9:31-5. [PMID: 16608393 DOI: 10.1089/rej.2006.9.31] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) show great promise for use in a variety of cell-based therapies. Because isolated primary mesenchymal stem cells are low in numbers, in vitro expansion is necessary. However, the expansion potential is limited and in vitro aging leads to loss of multipotency and replicative senescence. Stress induced by culture conditions is likely to be a major cause of replicative senescence and reduced multipotency of MSC and optimization of culture conditions might be able to reduce this. Caloric restriction (CR) is the only established method to delay aging and extend lifespan. In vitro caloric restriction experiments are rare, but have demonstrated beneficial effects. Therefore, we investigated the effect of culture medium glucose concentration on the proliferative and differentiation potential of mesenchymal stem cells. Reduction in glucose concentrations led to decreased apoptosis and an increased rate of MSC proliferation and increased the number and size of fibroblastic colonies in the colony-forming unit assay.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom.
| | | | | |
Collapse
|
41
|
Payushina OV, Domaratskaya EI, Starostin VI. Mesenchymal stem cells: Sources, phenotype, and differentiation potential. BIOL BULL+ 2006. [DOI: 10.1134/s106235900601002x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
42
|
Damrongsri D, Geva S, Salvi GE, Williams RC, Limwongse V, Offenbacher S. Cyclooxygenase-2 inhibition selectively attenuates bone morphogenetic protein-6 synthesis and bone formation during guided tissue regeneration in a rat model. Clin Oral Implants Res 2005; 17:38-47. [PMID: 16441783 DOI: 10.1111/j.1600-0501.2005.01187.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Bone formation during guided tissue regeneration is a tightly regulated process involving cells, extracellular matrix and growth factors. The aims of this study were (i) to examine the expression of cyclooxygenase-2 (COX-2) during bone regeneration and (ii) the effects of selective COX-2 inhibition on osseous regeneration and growth factor expression in the rodent femur model. MATERIAL AND METHODS A standardized transcortical defect of 5 x 1.5 mm was prepared in the femur of 12 male rats and a closed half-cylindrical titanium chamber was placed over the defect. The expression of COX-2 and of platelet-derived growth factor-B (PDGF-B), bone morphogenetic protein-6 (BMP-6) and insulin-like growth factor-I/II (IGF-I/II) was analyzed at Days 3, 7, 21 and 28 semiquantitatively by reverse transcriptase-polymerase chain reaction and immunohistochemistry. The effects of COX-2 inhibition by intraperitoneal injection of NS-398 (3 mg/kg/day) were analyzed in five additional animals sacrificed at Day 14. RESULTS Histomorphometry revealed that new bone formation occurred in the cortical defect area as well as in the supracortical region, i.e. region within the chamber by Day 7 and increased through Day 28. Immunohistochemical evidence of COX-2 and PDGF-B levels were observed early (i.e. Day 3) and decreased rapidly by Day 7. BMP-6 expression was maximal at Day 3 and slowly declined by Day 28. In contrast, IGF-I/II expression gradually increased during the 28-day period. Systemic administration NS-398 caused a statistically significant reduction (P<0.05) in new bone formation (25-30%) and was associated with a statistically significant reduction in BMP-6 protein and mRNA expression (50% and 65% at P<0.05 and P<0.01, respectively). PDGF-B mRNA or protein expression was not affected by NS-398 treatment. CONCLUSION COX-2 inhibition resulted in reduced BMP-6 expression and impaired osseous regeneration suggesting an important role for COX-2-induced signaling in BMP synthesis and new bone formation.
Collapse
Affiliation(s)
- Damrong Damrongsri
- Department of Anatomy, School of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
43
|
Chikazu D, Li X, Kawaguchi H, Sakuma Y, Voznesensky OS, Adams DJ, Xu M, Hoshi K, Katavic V, Herschman HR, Raisz LG, Pilbeam CC. Bone morphogenetic protein 2 induces cyclo-oxygenase 2 in osteoblasts via a Cbfa1 binding site: role in effects of bone morphogenetic protein 2 in vitro and in vivo. 2002. J Bone Miner Res 2005; 20:1888-98. [PMID: 16355502 DOI: 10.1359/jbmr.2005.20.10.1887] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
44
|
Vermelin L, Baroukh B, Llorens A, Saffar JL. Effects of essential fatty acid deficiency on periodontal tissue adaptation to spontaneous tooth migration. Calcif Tissue Int 2005; 77:30-6. [PMID: 16007482 DOI: 10.1007/s00223-004-0248-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2004] [Accepted: 01/21/2005] [Indexed: 11/24/2022]
Abstract
Essential fatty acids (EFAs) play a significant role in bone metabolism. Herein we studied the adaptation of alveolar bone to physiologic tooth drift in young rats deprived of essential fatty acids from birth. Reductions in femur size and trabecular bone volume reflected body growth impairment. Along the alveolar wall, osteoclastic resorption and bone formation were depressed, disrupting the adaptive deformation of the tooth socket to ongoing migration. As a result, the periodontal ligament narrowed considerably, and further adaptation was achieved through root resorption. Essential fatty acid deficiency (EFAD), did not affect precursor recruitment or differentiation in the periodontal ligament (PDL), but caused redistribution of nonspecific-esterase (NSE)-positive osteoclast precursors and tartrate-resistant acid phosphatase (TRAP)-positive pre-osteoclasts between the bone compartment (which was depleted) and the root compartment (which was enriched). EFAD had also a marked effect on the PDL vasculature; the number of vessels was reduced, whereas their size was markedly increased. As a whole, our results show that EFAD disturbs alveolar bone adaptation to drift, but that a reaction (detrimental to root integrity) prevents root collision with the bone surface, thereby preserving the PDL as a source of precursor cells for bone and cementum homeostasis. Moreover, our results confirm that although alveolar bone resorption is arachidonic acid-dependent, the factors activating root resorption are different.
Collapse
Affiliation(s)
- L Vermelin
- Laboratoire de Biologie et Physiopathologie Crânio-Faciales EA 2496, Faculté de Chirurgie Dentaire, Université René Descartes Paris-5, 1 rue M. Arnoux, 92120 Montrouge, France
| | | | | | | |
Collapse
|
45
|
Miao D, Murant S, Scutt N, Genever P, Scutt A. Megakaryocyte-bone marrow stromal cell aggregates demonstrate increased colony formation and alkaline phosphatase expression in vitro. ACTA ACUST UNITED AC 2005; 10:807-17. [PMID: 15265298 DOI: 10.1089/1076327041348473] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Bone marrow stromal cells (BMSCs) possess certain stem celllike properties and can differentiate to adopt a number of mesenchymal phenotypes. BMSCs are usually investigated in vitro as homogeneous single-cell suspensions; however, these preparations lose much of their osteogenic capacity. Using the fibroblastic colony-forming unit assay, we have compared the proliferation and capacity to express alkaline phosphatase of BMSC-containing aggregates of bone marrow cells with single-cell suspensions of bone marrow cells from the same source. Aggregates were separated from single cells by density gradient centrifugation or cell sieving. The aggregate and single-cell preparations gave rise to similar numbers of colonies; however, the colonies produced by the aggregates were larger and expressed higher levels of alkaline phosphatase. When the aggregates were dissociated, colonies still formed; however, they expressed negligible levels of alkaline phosphatase. Immunomagnetic selection and immunofluorescent staining for CD61, N-methyl-D-aspartate (NMDA) receptor subunit 1, and acetylcholinesterase showed that the majority of the aggregates giving rise to osteoblastic colonies contained megakaryocytes. These data demonstrate that removing BMSCs from their normal environment reduces their osteoblastic capacity and that to achieve their maximal differentiation, BMSCs require direct physical contact with accessory cells such as megakaryocytes. These findings may be of direct relevance to the use of BMSCs for tissue-engineering purposes.
Collapse
Affiliation(s)
- Dengshun Miao
- Department of Human Metabolism and Clinical Biochemistry, University of Sheffield, Sheffield, UK
| | | | | | | | | |
Collapse
|
46
|
Scutt A, Beier N, Fittschen C. EMD273316 & EMD95833, type 4 phosphodiesterase inhibitors, stimulate fibroblastic-colony formation by bone marrow cells via direct inhibition of PDE4 and the induction of endogenous prostaglandin synthesis. BMC Pharmacol 2004; 4:10. [PMID: 15219232 PMCID: PMC449707 DOI: 10.1186/1471-2210-4-10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Accepted: 06/25/2004] [Indexed: 11/10/2022] Open
Abstract
Background Type 4 phosphodiesterase (PDE4) inhibitors have been shown to stimulate bone formation in vivo and to stimulate osteoblastic differentiation in vitro. As one possible mechanism for the stimulation of bone formation is the recruitment of osteoprogenitor cells from the bone marrow, we have investigated the effect of the PDE4 inhibitors EMD273316, EMD95833, EMD249615 and EMD 219906 on fibroblastic colony formation by whole bone marrow cells and on the ability of these colonies to adopt an osteoblastic phenotype. Results All four agents stimulated colony formation in a concentration dependent manner, however, in the case of EMD273316 & EMD95833, the effect was evident at lower concentrations and the addition of prostaglandin E2 (PGE2) was not necessary for maximal stimulation. It was subsequently found that co-incubation with indomethacin reduced the stimulatory effects of EMD273316 & EMD95833 but had no effect on the actions of EMD249615 and EMD 219906 and that EMD273316 & EMD95833 stimulated the synthesis of endogenous PGE2 by whole bone marrow cells whereas EMD249615 and EMD 219906 had no significant effect. Conclusions These data suggest that EMD249615, EMD 219906, EMD273316 & EMD95833 can promote the recruitment of bone marrow osteoprogenitor cells leading to a stimulation of bone formation via their direct inhibitory effects on PDE4. The actions of EMD273316 & EMD95833 however, are augmented by their ability to stimulate endogenous prostanoids synthesis which acts synergistically with their direct effects on PDE4.
Collapse
Affiliation(s)
- Andrew Scutt
- Department of Engineering Materials and Sheffield Centre for Sports Medicine, University of Sheffield, Sheffield, UK
| | - Norbert Beier
- Merck KGaA, Biomedical Research, Diabetes and Complications, Darmstadt, Germany
| | - Claus Fittschen
- Merck KGaA, Biomedical Research, Immunology/Oncology, Darmstadt, Germany
| |
Collapse
|
47
|
Wang FS, Yang KD, Wang CJ, Huang HC, Chio CC, Hsu TY, Ou CY. Shockwave stimulates oxygen radical-mediated osteogenesis of the mesenchymal cells from human umbilical cord blood. J Bone Miner Res 2004; 19:973-82. [PMID: 15125794 DOI: 10.1359/jbmr.040121] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2003] [Revised: 10/29/2003] [Accepted: 01/16/2004] [Indexed: 11/18/2022]
Abstract
UNLABELLED Human umbilical cord blood (HUCB) mesenchymal progenitor cells expressed stro-1 or CD44 or CD29, and subsequently, differentiated toward osteogenic lineage. Physical shockwave treatment increased osteogenic activity of HUCB mesenchymal progenitor cells through superoxide-mediated TGF-beta1 induction. Transplantation of shockwave-treated HUCB mesenchymal progenitor cells enhanced healing of segmental femoral defect in severe combined immunodeficiency disease (SCID) mice. INTRODUCTION Mesenchymal progenitor cells (MPCs) in the bone marrow are precursors to bone development. It remains uncertain whether MPCs are present in human umbilical cord blood (HUCB) and are capable of differentiating into osteogenic cell lineage. Extending from a model of shockwave (SW) promotion of bone marrow stromal cell differentiation toward osteoprogenitors in rats, we further investigated how physical SW mediated biological responses in regulating osteogenic differentiation of HUCB MPCs. MATERIALS AND METHODS HUCB was subjected to SW treatment at different energy flux densities and impulses. Colony-forming units-stroma (CFU-Stroma), osteogenic activities (Cbfa1/Runx2 expression, bone alkaline phosphatase activity, and bone nodule formation), and bone formation by heterologous transplantation into SCID mice were assessed. RESULTS Few CD34+ stem cells (1.3%) and stro-1+ cells (1.0%) were present in the freshly prepared mononuclear cells (MNCs) from HUCB. The number of stro-1+ cells, but not CD34+, increased to 72.4% in the adherent cell culture over 6 days. Stro-1+ cells co-expressed CD44 and CD29 markers and grew into CFU-Stroma that matured into bone nodules. We found that the SW treatment (0.16 mJ/mm2 energy flux density, 200 impulses) elicited superoxide production and promoted formation of CFU-Stroma, but not of hematopoietic CFU-Mix. SW also enhanced the production of transforming growth factor (TGF)-beta1, but not of interleukin (IL)-3 or granulocyte monocyte-colony stimulating factor (GM-CSF). Neutralization of TGF-beta1 significantly reduced SW-promoted CFU-Stroma formation. Superoxide scavenging by superoxide dismutase blocked SW enhancement of TGF-beta1 production and formation of CFU-Stroma. Administration of SW-treated HUCB MPCs to SCID mice with femoral segmental defects facilitated dense, bridging callus and gap closure. CONCLUSION HUCB MPCs subjected to SW treatment is a potential source for stem cells useful in the treatment of orthopedic disorders. An optimal physical SW treatment enhanced osteogenesis through superoxide-mediated TGF-beta1 production. Physical stimulation is an alternative method for extending mesenchymal stem cells of HUCB.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Department of Medical Research, Chang Gung Memorial Hospital at Kaohsiung, Chang Gung University, Niau-Sung, Kaohsiung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
48
|
Weinreb M, Pollak RD, Ackerman Z. Experimental cholestatic liver disease through bile-duct ligation in rats results in skeletal fragility and impaired osteoblastogenesis. J Hepatol 2004; 40:385-90. [PMID: 15123350 DOI: 10.1016/j.jhep.2003.11.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2003] [Revised: 09/17/2003] [Accepted: 11/28/2003] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS Patients with cholestatic liver disease have 'low-turnover' osteoporosis. Since we reported that bile-duct ligated (BDL) rats develop bone disease with low bone formation and mass, we examined whether their reduced bone mass results in skeletal fragility, and whether the reduction in osteoprogenitor cells could explain the depressed bone formation. METHODS Four-week-old rats were pair-fed and subjected to BDL or sham surgery. After 4 weeks, ex vivo bone marrow stromal cell cultures were used to estimate the number of osteoprogenitors and tibial strength was measured by mechanical testing. The serum levels of albumin, bilirubin, alanine amino-transferase (ALT), alkaline phosphatase (ALP) and nitrite were measured. RESULTS BDL rats had elevated levels of bilirubin, ALT, ALP and nitrite. Tibiae of BDL rats were weaker than those of sham rats, exhibiting lower maximal force (-34%) and stiffness (-37%). The number of mineralized bone-like nodules in cultures from BDL rats was 65% lower than that in cultures from sham-operated rats, attesting to a diminished number of osteoprogenitors. CONCLUSIONS Skeletal fragility diminished osteoprogenitor pool and elevated plasma levels of nitrite are three additional characteristics of the bone disease that develops in BDL rats, thus increasing the validity of this animal model as representing the human bone disease in patients with cholestatic liver disease.
Collapse
Affiliation(s)
- Miron Weinreb
- Department of Oral Biology, Goldschleger School of Dental Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.
| | | | | |
Collapse
|
49
|
Shamir D, Keila S, Weinreb M. A selective EP4 receptor antagonist abrogates the stimulation of osteoblast recruitment from bone marrow stromal cells by prostaglandin E2 in vivo and in vitro. Bone 2004; 34:157-62. [PMID: 14751573 DOI: 10.1016/j.bone.2003.09.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Recent evidence indicates that systemic administration of PGE2 increases bone formation and bone mass via activation of the EP4 receptor. Previously, we demonstrated that osteoblastic recruitment from rat bone marrow stromal cells (BMSC) is a major mechanism for the anabolic effect of PGE2. In this study, we used a selective EP4 antagonist to test if the stimulation of osteoblast differentiation from rat BMSC in vitro and in vivo involves the EP4 receptor. In vitro, PGE2 (100 nM) increased nodule formation and alkaline phosphatase (ALP) activity in cultures of rat BMSC 1.5- to 2-fold. These effects were abolished by the EP4 antagonist at 10(-6) M but not 10(-9) M. Furthermore, PGE2 increased the number of surviving adherent BMSC by approximately 225% and the EP4 antagonist prevented this effect as well. The antagonist had no effect on basal levels of nodule formation and adherent cell number. In vivo, daily systemic administration of PGE2 at 6 mg/kg for 2 weeks increased cancellous bone area (by approximately 50%) and increased nodule formation (measured as mineralized area) in ex vivo stromal cultures by approximately 50%. Pre-administration of the EP4 antagonist at 10 mg/kg abrogated both the increase in bone mass as well as the increase in nodule formation. These data indicate that PGE2 stimulates osteoblastic commitment of BMSC via activation of the EP4 receptor.
Collapse
Affiliation(s)
- D Shamir
- Department of Oral Biology, Maurice and Gabriela Goldschleger School of Dental Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | | | | |
Collapse
|
50
|
Samoto H, Shimizu E, Matsuda-Honjyo Y, Saito R, Nakao S, Yamazaki M, Furuyama S, Sugiya H, Sodek J, Ogata Y. Prostaglandin E2 stimulates bone sialoprotein (BSP) expression through cAMP and fibroblast growth factor 2 response elements in the proximal promoter of the rat BSP gene. J Biol Chem 2003; 278:28659-67. [PMID: 12766167 DOI: 10.1074/jbc.m300671200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bone sialoprotein (BSP), an early marker of osteoblast differentiation, has been implicated in the nucleation of hydroxyapatite during de novo bone formation. Prostaglandin E2 (PGE2) has anabolic effects on proliferation and differentiation of osteoblasts via diverse signal transduction systems. Because PGE2 increases the proportion of functional osteoblasts in fetal rat calvarial cell cultures, we investigated the regulation of BSP, as an osteoblastic marker, by PGE2. Treatment of rat osteosarcoma UMR 106 cells with 3 microm, 300 nm, and 30 nm PGE2 increased the steady state levels of BSP mRNA about 2.7-, 2.5-, and 2.4-fold after 12 h. From transient transfection assays, the constructs including the promoter sequence of nucleotides (nt) -116 to +60 (pLUC3) were found to enhance transcriptional activity 3.8- and 2.2-fold treated with 3 microm and 30 nm PGE2 for 12 h. 2-bp mutations were made in an inverted CCAAT box (between nt -50 and -46), a cAMP response element (CRE; between nt -75 and -68), a fibroblast growth factor 2 response element (FRE; nt -92 to -85), and a pituitary-specific transcription factor-1 motif (between nt -111 and -105) within pLUC3 and pLUC7 constructs. Transcriptional stimulation by PGE2 was almost completed abrogated in constructs that included 2-bp mutations in either the CRE and FRE. In gel shift analyses an increased binding of nuclear extract components to double-stranded oligonucleotide probes containing CRE and FRE was observed following treatment with PGE2. These studies show that PGE2 induces BSP transcription in UMR 106 cells through juxtaposed CRE and FRE elements in the proximal promoter of the BSP gene.
Collapse
Affiliation(s)
- Hiroshi Samoto
- Periodontology, Endodontics, Pharmacology, Physiology, and Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|