1
|
Chatree K, Sriboonaied P, Phetkong C, Wattananit W, Chanchao C, Charoenpanich A. Distinctions in bone matrix nanostructure, composition, and formation between osteoblast-like cells, MG-63, and human mesenchymal stem cells, UE7T-13. Heliyon 2023; 9:e15556. [PMID: 37153435 PMCID: PMC10160763 DOI: 10.1016/j.heliyon.2023.e15556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 05/09/2023] Open
Abstract
Osteoblast-like cells and human mesenchymal stem cells (hMSCs) are frequently employed as osteoprogenitor cell models for evaluating novel biomaterials in bone healing and tissue engineering. In this study, the characterization of UE7T-13 hMSCs and MG-63 human osteoblast-like cells was examined. Both cells can undergo osteogenesis and produce calcium extracellular matrix; however, calcium nodules produced by MG-63 lacked a central mass and appeared flatter than UE7T-13. The absence of growing calcium nodules in MG-63 was discovered by SEM-EDX to be associated with the formation of alternating layers of cells and calcium extracellular matrix. The nanostructure and composition analysis showed that UE7T-13 had a finer nanostructure of calcium nodules with a higher calcium/phosphate ratio than MG-63. Both cells expressed high intrinsic levels of collagen type I alpha 1 chain, while only UE7T-13 expressed high levels of alkaline phosphatase, biomineralization associated (ALPL). High ALP activity in UE7T-13 was not further enhanced by osteogenic induction, but in MG-63, low intrinsic ALP activity was greatly induced by osteogenic induction. These findings highlight the differences between the two immortal osteoprogenitor cell lines, along with some technical notes that should be considered while selecting and interpreting the pertinent in vitro model.
Collapse
Affiliation(s)
- Kamonwan Chatree
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Patsawee Sriboonaied
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Chinnatam Phetkong
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Witoon Wattananit
- Scientific and Technological Equipment Centre, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Chanpen Chanchao
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Adisri Charoenpanich
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| |
Collapse
|
2
|
Lenna S, Brozovich A, Hirase T, Paradiso F, Weiner BK, Taraballi F. Comparison between Cancellous Trabecular and Cortical Specimens from Human Lumbar Spine Samples as an Alternative Source of Mesenchymal Stromal Cells. Stem Cells Dev 2022; 31:672-683. [PMID: 36039931 DOI: 10.1089/scd.2022.0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Due to their immunosuppressive potential and ability to differentiate into multiple musculoskeletal cell lineages, mesenchymal stromal cells (MSCs) became popular in clinical trials for the treatment of musculoskeletal disorders. The aim of this study was to isolate and characterize native populations of MSCs from human cortical and cancellous bone from the posterior elements of the lumbar spine and determine what source of MSCs yield better quality and quantity of cells to be potentially use for spinal fusion repair. We were able to show that MSCs from trabecular and cortical spine had the typical MSC morphology and expression markers; the ability to differentiate in adipocyte, chondrocyte, or osteoblast but they did not have a consistent pattern in the expression of the specific differentiation lineage genes. Moreover, MSCs from both sites demonstrated an immune suppression profile suggesting that these cells may have a more promising success in applications related to immunomodulation more than exploring their ability to drive osteogenesis to prevent nonunion in spine fusion procedures.
Collapse
Affiliation(s)
- Stefania Lenna
- Houston Methodist Research Institute, Houston, Texas, United States;
| | - Ava Brozovich
- Houston Methodist Academic Institute, Houston, Texas, United States;
| | - Takashi Hirase
- Houston Methodist Orthopedics & Sports Medicine Texas Medical Center, Houston, Texas, United States;
| | | | - Bradley K Weiner
- The Houston Methodist Research Institute, Department of Nanomedicine, Houston, Texas, United States.,Houston Methodist Hospital, Department of Orthopedic Surgery, Houston, Texas, United States;
| | | |
Collapse
|
3
|
Wang M, Xu X, Lei X, Tan J, Xie H. Mesenchymal stem cell-based therapy for burn wound healing. BURNS & TRAUMA 2021; 9:tkab002. [PMID: 34212055 PMCID: PMC8240555 DOI: 10.1093/burnst/tkab002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/14/2020] [Indexed: 02/05/2023]
Abstract
Burns, with their high incidence and mortality rates, have a devastating effect on patients. There are still huge challenges in the management of burns. Mesenchymal stem cells (MSCs), which have multidirectional differentiation potential, have aroused interest in exploring the capacity for treating different intractable diseases due to their strong proliferation, tissue repair, immune tolerance and paracrine abilities, among other features. Currently, several animal studies have shown that MSCs play various roles and have beneficial effects in promoting wound healing, inhibiting burn inflammation and preventing the formation of pathological scars during burn healing process. The substances MSCs secrete can act on peripheral cells and promote burn repair. According to preclinical research, MSC-based treatments can effectively improve burn wound healing and reduce pain. However, due to the small number of patients and the lack of controls, treatment plans and evaluation criteria vary widely, thus limiting the value of these clinical studies. Therefore, to better evaluate the safety and effectiveness of MSC-based burn treatments, standardization of the application scheme and evaluation criteria of MSC therapy in burn treatment is required in the future. In addition, the combination of MSC pretreatment and dressing materials are also conducive to improving the therapeutic effect of MSCs on burns. In this article, we review current animal research and clinical trials based on the use of stem cell therapy for treating burns and discuss the main challenges and coping strategies facing future clinical applications.
Collapse
Affiliation(s)
- Mingyao Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Xinxuan Xu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Xiongxin Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| |
Collapse
|
4
|
Cen YJ, You DB, Wang W, Feng Y. Preliminary studies of constructing a tissue-engineered lamellar corneal graft by culturing mesenchymal stem cells onto decellularized corneal matrix. Int J Ophthalmol 2021; 14:10-18. [PMID: 33469478 DOI: 10.18240/ijo.2021.01.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/28/2020] [Indexed: 12/28/2022] Open
Abstract
AIM To construct a competent corneal lamellar substitute in order to alleviate the shortage of human corneal donor. METHODS Rabbit mesenchymal stem cells (MSCs) were isolated from bone marrow and identified by flow cytometric, osteogenic and adipogenic induction. Xenogenic decellularized corneal matrix (XDCM) was generated from dog corneas. MSCs were seeded and cultured on XDCM to construct the tissue-engineered cornea. Post-transplantation biocompatibility of engineered corneal graft were tested by animal experiment. Rabbits were divided into two groups then underwent lamellar keratoplasty (LK) with different corneal grafts: 1) XDCM group (n=5): XDCM; 2) XDCM-MSCs groups (n=4): tissue-engineered cornea made up with XDCM and MSCs. The ocular surface recovery procedure was observed while corneal transparency, neovascularization and epithelium defection were measured and compared. In vivo on focal exam was performed 3mo postoperatively. RESULTS Rabbit MSCs were isolated and identified. Flow cytometry demonstrated isolated cells were CD90 positive and CD34, CD45 negative. Osteogenic and adipogenic induction verified their multipotent abilities. MSC-XDCM grafts were constructed and observed. In vivo transplantation showed the neovascularization in XDCM-MSC group was much less than that in XDCM group postoperatively. Post-transplant 3-month confocal test showed less nerve regeneration and bigger cell-absent area in XDCM-MSC group. CONCLUSION This study present a novel corneal tissue-engineered graft that could reduce post-operatively neovascularization and remain transparency, meanwhile shows that co-transplantation of MSCs may help increase corneal transplantation successful rate and enlarge the source range of corneal substitute to overcome cornea donor shortage.
Collapse
Affiliation(s)
- Yu-Jie Cen
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - De-Bo You
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wang
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
5
|
Anastasio A, Gergues M, Lebhar MS, Rameshwar P, Fernandez-Moure J. Isolation and characterization of mesenchymal stem cells in orthopaedics and the emergence of compact bone mesenchymal stem cells as a promising surgical adjunct. World J Stem Cells 2020; 12:1341-1353. [PMID: 33312402 PMCID: PMC7705465 DOI: 10.4252/wjsc.v12.i11.1341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/26/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
The potential clinical and economic impact of mesenchymal stem cell (MSC) therapy is immense. MSCs act through multiple pathways: (1) as “trophic” cells, secreting various factors that are immunomodulatory, anti-inflammatory, anti-apoptotic, proangiogenic, proliferative, and chemoattractive; (2) in conjunction with cells native to the tissue they reside in to enhance differentiation of surrounding cells to facilitate tissue regrowth. Researchers have developed methods for the extraction and expansion of MSCs from animal and human tissues. While many sources of MSCs exist, including adipose tissue and iliac crest bone graft, compact bone (CB) MSCs have shown great potential for use in orthopaedic surgery. CB MSCs exert powerful immunomodulatory effects in addition to demonstrating excellent regenerative capacity for use in filling boney defects. CB MSCs have been shown to have enhanced response to hypoxic conditions when compared with other forms of MSCs. More work is needed to continue to characterize the potential applications for CB MSCs in orthopaedic trauma.
Collapse
Affiliation(s)
- Albert Anastasio
- Department of Orthopedic Surgery, Duke University Health System, Durham, NC 27710, United States
| | - Marina Gergues
- Department of Medicine, Hematology/Oncology, Rutgers University, New Jersey Medical School, Newark, NJ 07103, United States
| | - Michael S Lebhar
- School of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Pranela Rameshwar
- Department of Medicine-Hematology/Oncology, Rutgers School of Biomedical Health Science, Newark, NJ 07103, United States
| | - Joseph Fernandez-Moure
- Department of Surgery, Division of Trauma, Acute, and Critical Care Surgery, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
6
|
Chougule A, Kolli V, Baroi S, Ebraheim N, Czernik PJ, Loh YP, Lecka-Czernik B. Nonenzymatic and Trophic Activities of Carboxypeptidase E Regulate Bone Mass and Bioenergetics of Skeletal Stem Cells in Mice. JBMR Plus 2020; 4:e10392. [PMID: 32995694 PMCID: PMC7507073 DOI: 10.1002/jbm4.10392] [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: 04/16/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/20/2022] Open
Abstract
Bone and energy metabolism are integrated by common regulatory mechanisms. Carboxypeptidase E (CPE), also known as obesity susceptibility protein or neurotrophic factor‐α1, is recognized for its function in processing prohormones, including proinsulin and pro‐opiomelanocortin polypeptide. Independent of its enzymatic activity, CPE may also act as a secreted factor with divergent roles in neuroprotection and cancer growth; however, its role in the regulation of bone mass and skeletal cell differentiation is unknown. Male mice with global deficiency in CPE are characterized with profound visceral obesity, low bone mass in both appendicular and axial skeleton, and high volume of marrow fat. Interestingly, although metabolic deficit of CPE KO mice develops early in life, bone deficit develops in older age, suggesting that CPE bone‐specific activities differ from its enzymatic activities. Indeed, mutated CPE knockin (mCPE KI) mice ectopically expressing CPE‐E342Q, a mutated protein lacking enzymatic activity, develop the same obese phenotype and accumulate the same volume of marrow fat as CPE KO mice, but their bone mass is normal. In addition, differentiation of marrow hematopoietic cells toward tartrate‐resistant acid phosphatase‐positive multinucleated osteoclasts is highly increased in CPE KO mice, but normal in mCPE KI mice. Moreover, in murine skeletal stem cells, nonenzymatic trophic CPE has activated ERK signaling, increased cell proliferation and increased mitochondrial activity. Treatment of preosteoblastic cells with intact or mutated recombinant CPE led to a transient accumulation of small lipid droplets, increased oxidative phosphorylation, and increased cellular dependence on fatty acids as fuel for energy production. In human marrow aspirates, CPE expression increases up to 30‐fold in osteogenic conditions. These findings suggest that nonenzymatic and trophic activities of CPE regulate bone mass, whereas marrow adiposity is controlled by CPE enzymatic activity. Thus, CPE can be positioned as a factor regulating simultaneously bone and energy metabolism through a combination of shared and distinct mechanisms. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Amit Chougule
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Vipula Kolli
- Section on Cellular Neurobiology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health Bethesda MD USA
| | - Sudipta Baroi
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Nabil Ebraheim
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Piotr J Czernik
- Department of Physiology and Pharmacology University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Y Peng Loh
- Section on Cellular Neurobiology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health Bethesda MD USA
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Department of Physiology and Pharmacology University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| |
Collapse
|
7
|
Li K, Ning T, Wang H, Jiang Y, Zhang J, Ge Z. Nanosecond pulsed electric fields enhance mesenchymal stem cells differentiation via DNMT1-regulated OCT4/NANOG gene expression. Stem Cell Res Ther 2020; 11:308. [PMID: 32698858 PMCID: PMC7374836 DOI: 10.1186/s13287-020-01821-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 02/28/2023] Open
Abstract
Background Multiple strategies have been proposed to promote the differentiation potential of mesenchymal stem cells (MSCs), which is the fundamental property in tissue formation and regeneration. However, these strategies are relatively inefficient that limit the application. In this study, we reported a novel and efficient strategy, nanosecond pulsed electric fields (nsPEFs) stimulation, which can enhance the trilineage differentiation potential of MSCs, and further explained the mechanism behind. Methods We used histological staining to screen out the nsPEFs parameters that promoted the trilineage differentiation potential of MSCs, and further proved the effect of nsPEFs by detecting the functional genes. In order to explore the corresponding mechanism, we examined the expression of pluripotency genes and the methylation status of their promoters. Finally, we targeted the DNA methyltransferase which was affected by nsPEFs. Results The trilineage differentiation of bone marrow-derived MSCs was significantly enhanced in vitro by simply pre-treating with 5 pulses of nsPEFs stimulation (energy levels as 10 ns, 20 kV/cm; 100 ns, 10 kV/cm), due to that the nsPEFs demethylated the promoters of stem cell pluripotency genes OCT4 and NANOG through instantaneous downregulation of DNA methylation transferase 1 (DNMT1), thereby increasing the expression of OCT4 and NANOG for up to 3 days, and created a treatment window period of stem cells. Conclusions In summary, nsPEFs can enhance MSCs differentiation via the epigenetic regulation and could be a safe and effective strategy for future stem cell application.
Collapse
Affiliation(s)
- Kejia Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Tong Ning
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Hao Wang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jue Zhang
- Institute of Biomechanics and Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
Skeletal-muscle-derived mesenchymal stem/stromal cells from patients with osteoarthritis show superior biological properties compared to bone-derived cells. Stem Cell Res 2019; 38:101465. [PMID: 31132579 DOI: 10.1016/j.scr.2019.101465] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are being exploited for patient-derived stem-cell therapies. As the biological properties of MSCs derived from skeletal muscle of osteoarthritis patients are poorly understood, the aim of this study was to compare muscle MSCs with well-recognized bone and bone marrow-derived MSCs from these patients. Paired samples of skeletal muscle and trabecular bone tissue were obtained from 21 patients with osteoarthritis. Isolated cells were compared using ex vivo immunophenotyping and detailed in vitro analyses. These included the colony forming unit fibroblast assay, growth kinetics, senescence, multilineage potential, immunophenotyping, and MSC marker gene expression profiling. Freshly isolated MSCs from muscle showed improved viability over bone-derived MSCs, with similar mesenchymal fraction. Muscle-derived MSCs showed superior clonogenicity, higher growth rates, and lower doubling times. Muscle-derived MSCs also showed superior osteogenic and myogenic properties and a positive correlation between CD271 expression and adipogenesis. Senescence rate as well as adipogenic and chondrogenic potentials were similar. Skeletal muscle-derived MSCs of osteoarthritis patients have superior clonogenicity and growth kinetics compared to bone-derived MSCs, making them a good candidate for autologous stem-cell therapies. Moreover, the positive correlation between CD271 and adipogenesis suggest that CD271 expressing muscle MSCs might contribute to muscle steatosis observed in osteoarthritis.
Collapse
|
9
|
González-Gil AB, Lamo-Espinosa JM, Muiños-López E, Ripalda-Cemboráin P, Abizanda G, Valdés-Fernández J, López-Martínez T, Flandes-Iparraguirre M, Andreu I, Elizalde MR, Stuckensen K, Groll J, De-Juan-Pardo EM, Prósper F, Granero-Moltó F. Periosteum-derived mesenchymal progenitor cells in engineered implants promote fracture healing in a critical-size defect rat model. J Tissue Eng Regen Med 2019; 13:742-752. [PMID: 30785671 DOI: 10.1002/term.2821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/01/2019] [Accepted: 02/13/2019] [Indexed: 11/06/2022]
Abstract
An attractive alternative to bone autografts is the use of autologous mesenchymal progenitor cells (MSCs) in combination with biomaterials. We compared the therapeutic potential of different sources of mesenchymal stem cells in combination with biomaterials in a bone nonunion model. A critical-size defect was created in Sprague-Dawley rats. Animals were divided into six groups, depending on the treatment to be applied: bone defect was left empty (CTL); treated with live bone allograft (LBA); hrBMP-2 in collagen scaffold (CSBMP2 ); acellular polycaprolactone scaffold (PCL group); PCL scaffold containing periosteum-derived MSCs (PCLPMSCs ) and PCL containing bone marrow-derived MSCs (PCLBMSCs ). To facilitate cell tracking, both MSCs and bone graft were isolated from green fluorescent protein (GFP)-transgenic rats. CTL group did not show any signs of healing during the radiological follow-up (n = 6). In the LBA group, all the animals showed bone bridging (n = 6) whereas in the CSBMP2 group, four out of six animals demonstrated healing. In PCL and PCLPMSCs groups, a reduced number of animals showed radiological healing, whereas no healing was detected in the PCLBMSCs group. Using microcomputed tomography, the bone volume filling the defect was quantified, showing significant new bone formation in the LBA, CSBMP2 , and PCLPMSCs groups when compared with the CTL group. At 10 weeks, GFP positive cells were detected only in the LBA group and restricted to the outer cortical bone in close contact with the periosteum. Tracking of cellular implants demonstrated significant survival of the PMSCs when compared with BMSCs. In conclusion, PMSCs improve bone regeneration being suitable for mimetic autograft design.
Collapse
Affiliation(s)
- Ana B González-Gil
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - José M Lamo-Espinosa
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Emma Muiños-López
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Gloria Abizanda
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - José Valdés-Fernández
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Tania López-Martínez
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Ion Andreu
- TECNUN, Universidad de Navarra, San Sebastian, Spain
| | - María Reyes Elizalde
- TECNUN, Universidad de Navarra, San Sebastian, Spain.,CEIT, San Sebastian, Spain
| | - Kai Stuckensen
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, Germany
| | - Elena M De-Juan-Pardo
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Felipe Prósper
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain.,Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Froilán Granero-Moltó
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
10
|
Mao L, Wang M, Xi X, Dai Y, Wang N, Wang J, Xue C. Sialoglycoprotein isolated from Carassius auratus eggs promotes osteogenesis by stimulating mesenchymal stem cells to commit to osteoblast differentiation. Cell Tissue Res 2019; 376:365-376. [PMID: 30715559 DOI: 10.1007/s00441-018-2976-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
In this study, we explore whether the pro-osteogenic effects of sialoglycoprotein from Carassius auratus eggs (Ca-SGP) involve mesenchymal stem cells (MSCs). Ovariectomized osteoporotic mice treated with Ca-SGP had increased bone formation and reduced bone marrow adipose tissue. As MSCs are common progenitors of osteoblasts and adipocytes, we isolated MSCs from Ca-SGP-treated mice and found that they tended to differentiate into osteoblasts over adipocytes confirmed by Alizarin red and Oil red O staining. This change was seen at the gene and protein level. To further explore the effect of Ca-SGP on MSCs, we isolated MSCs from healthy mice and treated them with Ca-SGP in vitro. We discovered that Ca-SGP promoted MSC differentiation to osteoblasts. In addition, Ca-SGP promoted osteogenesis and reduced the fat in marrow cavity of adolescent mice. For the first time, our results demonstrate that Ca-SGP promotes osteogenesis via stimulating MSCs to commit to osteoblasts. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Lei Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Meiling Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Xingjun Xi
- China National Institute of Standardization, Beijing, 100191, China
| | - Yufeng Dai
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Na Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China.
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| |
Collapse
|
11
|
Basic Science and Clinical Application of Reamed Sources for Autogenous Bone Graft Harvest. J Am Acad Orthop Surg 2018; 26:420-428. [PMID: 29781821 DOI: 10.5435/jaaos-d-16-00512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Autologous bone graft remains the only clinically available source of graft material with osteogenic, osteoinductive, and osteoconductive properties. Although iliac crest autologous bone graft has long served as the benchmark, reamed autogenous bone graft offers several advantages. Reamed autograft has a biochemical and cellular profile that is at least equivalent, and perhaps superior, to that of iliac crest autograft. In addition, larger volumes of reamed autograft can be obtained via less-invasive techniques, giving surgeons an accessible source of mesenchymal stem cells that can be reliably and repeatedly harvested. Early clinical experience involving reamed autogenous bone graft in the management of nonunion, bone defects, and arthrodesis has been encouraging and has demonstrated the necessary properties to warrant regular consideration of reamed graft for these applications.
Collapse
|
12
|
Čamernik K, Barlič A, Drobnič M, Marc J, Jeras M, Zupan J. Mesenchymal Stem Cells in the Musculoskeletal System: From Animal Models to Human Tissue Regeneration? Stem Cell Rev Rep 2018; 14:346-369. [DOI: 10.1007/s12015-018-9800-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
Christou I, Mallis P, Michalopoulos E, Chatzistamatiou T, Mermelekas G, Zoidakis J, Vlahou A, Stavropoulos-Giokas C. Evaluation of Peripheral Blood and Cord Blood Platelet Lysates in Isolation and Expansion of Multipotent Mesenchymal Stromal Cells. Bioengineering (Basel) 2018; 5:19. [PMID: 29495420 PMCID: PMC5874885 DOI: 10.3390/bioengineering5010019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/16/2018] [Accepted: 02/24/2018] [Indexed: 02/07/2023] Open
Abstract
Abstract: Background: Multipotent Mesenchymal Stromal Cells (MSCs) are used in tissue engineering and regenerative medicine. The in vitro isolation and expansion of MSCs involve the use of foetal bovine serum (FBS). However, many concerns have been raised regarding the safety of this product. In this study, alternative additives derived either from peripheral or cord blood were tested as an FBS replacement. Methods: Platelet lysates (PL) from peripheral and cord blood were used for the expansion of MSCs. The levels of growth factors in peripheral blood (PB) and cord blood (CB) PLs were determined using the Multiple Reaction Monitoring (MRM). Finally, the cell doubling time (CDT), tri-lineage differentiation and phenotypic characterization of the MSCs expanded with FBS and PLs were determined. Results: MSCs treated with culture media containing FBS and PB-PL, were successfully isolated and expanded, whereas MSCs treated with CB-PL could not be maintained in culture. Furthermore, the MRM analysis yielded differences in growth factor levels between PB-PL and CB-PL. In addition, the MSCs were successfully expanded with FBS and PB-PL and exhibited tri-lineage differentiation and stable phenotypic characteristics. Conclusion: PB-PL could be used as an alternative additive for the production of MSCs culture medium applied to xenogeneic-free expansion and maintenance of MSCs in large scale clinical studies.
Collapse
Affiliation(s)
- Ioanna Christou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Theofanis Chatzistamatiou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - George Mermelekas
- Biotechnology division, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Jerome Zoidakis
- Biotechnology division, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Antonia Vlahou
- Biotechnology division, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece.
| |
Collapse
|
14
|
Le BQ, Nurcombe V, Cool SM, van Blitterswijk CA, de Boer J, LaPointe VLS. The Components of Bone and What They Can Teach Us about Regeneration. MATERIALS (BASEL, SWITZERLAND) 2017; 11:E14. [PMID: 29271933 PMCID: PMC5793512 DOI: 10.3390/ma11010014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022]
Abstract
The problem of bone regeneration has engaged both physicians and scientists since the beginning of medicine. Not only can bone heal itself following most injuries, but when it does, the regenerated tissue is often indistinguishable from healthy bone. Problems arise, however, when bone does not heal properly, or when new tissue is needed, such as when two vertebrae are required to fuse to stabilize adjacent spine segments. Despite centuries of research, such procedures still require improved therapeutic methods to be devised. Autologous bone harvesting and grafting is currently still the accepted benchmark, despite drawbacks for clinicians and patients that include limited amounts, donor site morbidity, and variable quality. The necessity for an alternative to this "gold standard" has given rise to a bone-graft and substitute industry, with its central conundrum: what is the best way to regenerate bone? In this review, we dissect bone anatomy to summarize our current understanding of its constituents. We then look at how various components have been employed to improve bone regeneration. Evolving strategies for bone regeneration are then considered.
Collapse
Affiliation(s)
- Bach Quang Le
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
| | - Simon McKenzie Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore 119288, Singapore.
| | - Clemens A van Blitterswijk
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Jan de Boer
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Vanessa Lydia Simone LaPointe
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
15
|
Fellows CR, Matta C, Zakany R, Khan IM, Mobasheri A. Adipose, Bone Marrow and Synovial Joint-Derived Mesenchymal Stem Cells for Cartilage Repair. Front Genet 2016; 7:213. [PMID: 28066501 PMCID: PMC5167763 DOI: 10.3389/fgene.2016.00213] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/22/2016] [Indexed: 01/15/2023] Open
Abstract
Current cell-based repair strategies have proven unsuccessful for treating cartilage defects and osteoarthritic lesions, consequently advances in innovative therapeutics are required and mesenchymal stem cell-based (MSC) therapies are an expanding area of investigation. MSCs are capable of differentiating into multiple cell lineages and exerting paracrine effects. Due to their easy isolation, expansion, and low immunogenicity, MSCs are an attractive option for regenerative medicine for joint repair. Recent studies have identified several MSC tissue reservoirs including in adipose tissue, bone marrow, cartilage, periosteum, and muscle. MSCs isolated from these discrete tissue niches exhibit distinct biological activities, and have enhanced regenerative potentials for different tissue types. Each MSC type has advantages and disadvantages for cartilage repair and their use in a clinical setting is a balance between expediency and effectiveness. In this review we explore the challenges associated with cartilage repair and regeneration using MSC-based cell therapies and provide an overview of phenotype, biological activities, and functional properties for each MSC population. This paper also specifically explores the therapeutic potential of each type of MSC, particularly focusing on which cells are capable of producing stratified hyaline-like articular cartilage regeneration. Finally we highlight areas for future investigation. Given that patients present with a variety of problems it is unlikely that cartilage regeneration will be a simple "one size fits all," but more likely an array of solutions that need to be applied systematically to achieve regeneration of a biomechanically competent repair tissue.
Collapse
Affiliation(s)
| | - Csaba Matta
- Faculty of Health and Medical Sciences, University of SurreyGuildford, UK
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecen, Hungary
| | - Roza Zakany
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecen, Hungary
| | - Ilyas M. Khan
- Centre for NanoHealth, Swansea University Medical SchoolSwansea, UK
| | - Ali Mobasheri
- Faculty of Health and Medical Sciences, University of SurreyGuildford, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical CentreNottingham, UK
- King Fahd Medical Research Center, King AbdulAziz UniversityJeddah, Saudi Arabia
- Sheik Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King AbdulAziz UniversityJeddah, Saudi Arabia
| |
Collapse
|
16
|
Fenger JM, Roberts RD, Iwenofu OH, Bear MD, Zhang X, Couto JI, Modiano JF, Kisseberth WC, London CA. MiR-9 is overexpressed in spontaneous canine osteosarcoma and promotes a metastatic phenotype including invasion and migration in osteoblasts and osteosarcoma cell lines. BMC Cancer 2016; 16:784. [PMID: 27724924 PMCID: PMC5057229 DOI: 10.1186/s12885-016-2837-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 10/05/2016] [Indexed: 01/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) regulate the expression of networks of genes and their dysregulation is well documented in human malignancies; however, limited information exists regarding the impact of miRNAs on the development and progression of osteosarcoma (OS). Canine OS exhibits clinical and molecular features that closely resemble the corresponding human disease and it is considered a well-established spontaneous animal model to study OS biology. The purpose of this study was to investigate miRNA dysregulation in canine OS. Methods We evaluated miRNA expression in primary canine OS tumors and normal canine osteoblast cells using the nanoString nCounter system. Quantitative PCR was used to validate the nanoString findings and to assess miR-9 expression in canine OS tumors, OS cell lines, and normal osteoblasts. Canine osteoblasts and OS cell lines were stably transduced with pre-miR-9 or anti-miR-9 lentiviral constructs to determine the consequences of miR-9 on cell proliferation, apoptosis, invasion and migration. Proteomic and gene expression profiling of normal canine osteoblasts with enforced miR-9 expression was performed using 2D-DIGE/tandem mass spectrometry and RNA sequencing and changes in protein and mRNA expression were validated with Western blotting and quantitative PCR. OS cell lines were transduced with gelsolin (GSN) shRNAs to investigate the impact of GSN knockdown on OS cell invasion. Results We identified a unique miRNA signature associated with primary canine OS and identified miR-9 as being significantly overexpressed in canine OS tumors and cell lines compared to normal osteoblasts. Additionally, high miR-9 expression was demonstrated in tumor-specific tissue obtained from primary OS tumors. In normal osteoblasts and OS cell lines transduced with miR-9 lentivirus, enhanced invasion and migration were observed, but miR-9 did not affect cell proliferation or apoptosis. Proteomic and transcriptional profiling of normal canine osteoblasts overexpressing miR-9 identified alterations in numerous genes, including upregulation of GSN, an actin filament-severing protein involved in cytoskeletal remodeling. Lastly, stable downregulation of miR-9 in OS cell lines reduced GSN expression with a concomitant decrease in cell invasion and migration; concordantly, cells transduced with GSN shRNA demonstrated decreased invasive properties. Conclusions Our findings demonstrate that miR-9 promotes a metastatic phenotype in normal canine osteoblasts and malignant OS cell lines, and that this is mediated in part by enhanced GSN expression. As such, miR-9 represents a novel target for therapeutic intervention in OS. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2837-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joelle M Fenger
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon L. Tharp Street, Columbus, OH, USA. .,, 444 Veterinary Medical Academic Building, 1600 Coffey Road, Columbus, OH, 43210, USA.
| | - Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
| | - O Hans Iwenofu
- Department of Pathology, College of Medicine, The Ohio State University, 129 Hamilton Hall, 1645 Neil Avenue, Columbus, OH, USA
| | - Misty D Bear
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1900 Coffey Road, Columbus, OH, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Drive, Columbus, OH, USA
| | - Jason I Couto
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon L. Tharp Street, Columbus, OH, USA
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA.,Masonic Cancer Center, University of Minnesota, 420 Delaware Street, SE, MMC 806, Minneapolis, MN, USA
| | - William C Kisseberth
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon L. Tharp Street, Columbus, OH, USA
| | - Cheryl A London
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon L. Tharp Street, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1900 Coffey Road, Columbus, OH, USA
| |
Collapse
|
17
|
Mesenchymal Stem/Stromal Cells from Discarded Neonatal Sternal Tissue: In Vitro Characterization and Angiogenic Properties. Stem Cells Int 2015; 2016:5098747. [PMID: 26770206 PMCID: PMC4684890 DOI: 10.1155/2016/5098747] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
Autologous and nonautologous bone marrow mesenchymal stem/stromal cells (MSCs) are being evaluated as proangiogenic agents for ischemic and vascular disease in adults but not in children. A significant number of newborns and infants with critical congenital heart disease who undergo cardiac surgery already have or are at risk of developing conditions related to inadequate tissue perfusion. During neonatal cardiac surgery, a small amount of sternal tissue is usually discarded. Here we demonstrate that MSCs can be isolated from human neonatal sternal tissue using a nonenzymatic explant culture method. Neonatal sternal bone MSCs (sbMSCs) were clonogenic, had a surface marker expression profile that was characteristic of bone marrow MSCs, were multipotent, and expressed pluripotency-related genes at low levels. Neonatal sbMSCs also demonstrated in vitro proangiogenic properties. Sternal bone MSCs cooperated with human umbilical vein endothelial cells (HUVECs) to form 3D networks and tubes in vitro. Conditioned media from sbMSCs cultured in hypoxia also promoted HUVEC survival and migration. Given the neonatal source, ease of isolation, and proangiogenic properties, sbMSCs may have relevance to therapeutic applications.
Collapse
|
18
|
Che X, Guo J, Li X, Wang L, Wei S. Intramuscular injection of bone marrow mononuclear cells contributes to bone repair following midpalatal expansion in rats. Mol Med Rep 2015; 13:681-8. [PMID: 26648442 PMCID: PMC4686095 DOI: 10.3892/mmr.2015.4578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 10/19/2015] [Indexed: 12/21/2022] Open
Abstract
Healing from injury requires the activation and proliferation of stem cells for tissue repair. Previous studies have demonstrated that bone marrow is a central pool of stem cells. The present study aimed to investigate the route undertaken by bone marrow mononuclear cells (BMMCs) following BMMC transplantation by masseter injection in a rat model of midpalatal expansion. The rats were divided into five groups according to the types of midpalatal expansion, incision and BMMC transplantation. Samples of midpalatal bone from the rats in each group were used for histological and immunohistochemical assessments to track and evaluate the differential potentials of the transplanted BMMCs in the masseter muscle and midpalatal bone. Bromodeoxyuridine was used as a BMMC tracing label, and M-cadherin was used to detect muscle satellite cells. The BMMCs injected into the masseter were observed, not only in the masseter, but also in the blood vessels and oral mucosa, and enveloped the midpalatal bone. A number of the BMMCs transformed into osteoblasts at the boundary of the neuromuscular bundle, and were embedded in the newly formed bone during midpalatal bone regeneration. The results of the present study suggested that BMMCs entered the circulation and migrated from muscle to the bone tissue, where they were involved in bone repair. Therefore, BMMCs may prove useful in the treatment of various types of cancer.
Collapse
Affiliation(s)
- Xiaoxia Che
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, P.R. China
| | - Jie Guo
- Department of Orthodontics, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Biomedicine, Jinan, Shandong 250012, P.R. China
| | - Xiangdong Li
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, P.R. China
| | - Lve Wang
- Department of Microbiology, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Silong Wei
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
19
|
Fernandez-Moure JS, Corradetti B, Chan P, Van Eps JL, Janecek T, Rameshwar P, Weiner BK, Tasciotti E. Enhanced osteogenic potential of mesenchymal stem cells from cortical bone: a comparative analysis. Stem Cell Res Ther 2015; 6:203. [PMID: 26503337 PMCID: PMC4620594 DOI: 10.1186/s13287-015-0193-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/20/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) hold great promise for regenerative therapies in the musculoskeletal system. Although MSCs from bone marrow (BM-MSCs) and adipose tissue (AD-MSCs) have been extensively characterized, there is still debate as to the ideal source of MSCs for tissue-engineering applications in bone repair. Methods MSCs were isolated from cortical bone fragments (CBF-MSCs) obtained from patients undergoing laminectomy, selected by fluorescence-activated cell sorting analysis, and tested for their potential to undergo mesodermic differentiation. CBF-MSCs were then compared with BM-MSCs and AD-MSCs for their colony-forming unit capability and osteogenic potential in both normoxia and hypoxia. After 2 and 4 weeks in inducing media, differentiation was assessed qualitatively and quantitatively by the evaluation of alkaline phosphatase (ALP) expression and mineral deposition (Von Kossa staining). Transcriptional activity of osteoblastogenesis-associated genes (Alp, RUNX2, Spp1, and Bglap) was also analyzed. Results The cortical fraction of the bone contains a subset of cells positive for MSC-associated markers and capable of tri-lineage differentiation. The hypoxic conditions were generally more effective in inducing osteogenesis for the three cell lines. However, at 2 and 4 weeks, greater calcium deposition and ALP expression were observed in both hypoxic and normoxic conditions in CBF-MSCs compared with AD- and BM-MSCs. These functional observations were further corroborated by gene expression analysis, which showed a significant upregulation of Bglap, Alp, and Spp1, with a 22.50 (±4.55)-, 46.56 (±7.4)-, 71.46 (±4.16)-fold increase compared with their uninduced counterparts. Conclusions This novel population of MSCs retains a greater biosynthetic activity in vitro, which was found increased in hypoxic conditions. The present study demonstrates that quantitative differences between MSCs retrieved from bone marrow, adipose, and the cortical portion of the bone with respect to their osteogenic potential exist and suggests the cortical bone as suitable candidate to use for orthopedic tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Joseph S Fernandez-Moure
- Houston Methodist Hospital Department of Surgery, Houston, USA. .,Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
| | - Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA. .,Department of Life and Environmental Sciences, Università Politecnica delle Marche, via Brecce Bianche, 60131, Ancona, Italy.
| | - Paige Chan
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
| | - Jeffrey L Van Eps
- Houston Methodist Hospital Department of Surgery, Houston, USA. .,Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
| | - Trevor Janecek
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ, 07103, USA.
| | - Bradley K Weiner
- Houston Methodist Hospital Department of Orthopedic Surgery, 6565 Fannin Street, Houston, TX, 77030, USA.
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
| |
Collapse
|
20
|
Corradetti B, Taraballi F, Powell S, Sung D, Minardi S, Ferrari M, Weiner BK, Tasciotti E. Osteoprogenitor cells from bone marrow and cortical bone: understanding how the environment affects their fate. Stem Cells Dev 2015; 24:1112-23. [PMID: 25517215 DOI: 10.1089/scd.2014.0351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone is a dynamic organ where skeletal progenitors and hematopoietic cells share and compete for space. Presumptive mesenchymal stem cells (MSC) have been identified and harvested from the bone marrow (BM-MSC) and cortical bone fragments (CBF-MSC). In this study, we demonstrate that despite the cells sharing a common ancestor, the differences in the structural properties of the resident tissues affect cell behavior and prime them to react differently to stimuli. Similarly to the bone marrow, the cortical portion of the bone contains a unique subset of cells that stains positively for the common MSC-associated markers. These cells display different multipotent differentiation capability, clonogenic expansion, and immunosuppressive potential. In particular, when compared with BM-MSC, CBF-MSC are bigger in size, show a lower proliferation rate at early passages, have a greater commitment toward the osteogenic lineage, constitutively produce nitric oxide as a mediator for bone remodeling, and more readily respond to proinflammatory cytokines. Our data suggest that the effect of the tissue's microenvironment makes the CBF-MSC a superior candidate in the development of new strategies for bone repair.
Collapse
Affiliation(s)
- Bruna Corradetti
- 1 Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhang Y, Zhang H, Zhang G, Ka K, Huang W. Combining acellular nerve allografts with brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells restores sciatic nerve injury better than either intervention alone. Neural Regen Res 2014; 9:1814-9. [PMID: 25422643 PMCID: PMC4239771 DOI: 10.4103/1673-5374.143427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2014] [Indexed: 01/08/2023] Open
Abstract
In this study, we chemically extracted acellular nerve allografts from bilateral sciatic nerves, and repaired 10-mm sciatic nerve defects in rats using these grafts and brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells. Experiments were performed in three groups: the acellular nerve allograft bridging group, acellular nerve allograft + bone marrow mesenchymal stem cells group, and the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group. Results showed that at 8 weeks after bridging, sciatic functional index, triceps wet weight recovery rate, myelin thickness, and number of myelinated nerve fibers were significantly changed in the three groups. Variations were the largest in the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group compared with the other two groups. Experimental findings suggest that chemically extracted acellular nerve allograft combined nerve factor and mesenchymal stem cells can promote the restoration of sciatic nerve defects. The repair effect seen is better than the single application of acellular nerve allograft or acellular nerve allograft combined mesenchymal stem cell transplantation.
Collapse
Affiliation(s)
- Yanru Zhang
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China ; Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Gechen Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ka Ka
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenhua Huang
- Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
22
|
Conway JD, Shabtai L, Specht SC, Herzenberg JE. Sequential harvesting of bone graft from the intramedullary canal of the femur. Orthopedics 2014; 37:e796-803. [PMID: 25350622 DOI: 10.3928/01477447-20140825-56] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/30/2014] [Indexed: 02/03/2023]
Abstract
The effectiveness of using the Reamer/Irrigator/Aspirator (RIA) System (Synthes, Inc, West Chester, Pennsylvania) to obtain bone graft from the intramedullary canal of long bones for the treatment of bone defects and nonunions has been previously documented. However, there is nothing in the literature discussing the potential for reaming the same canal at subsequent surgeries. The authors detail their experience of 8 instances of sequential reaming in 7 patients. Six patients were harvested twice, and 1 patient was harvested 3 times. In each patient, the bone graft was obtained from the same canal. The main outcome measurements were time interval between reamings, reamer head size, indication for reaming, volume of harvested bone graft, and complications. Average volume of graft obtained in the first reaming procedure was 34 mL (range, 25-50 mL). After an average of 9 months (range, 3-16 months), the subsequent reaming was performed. Average volume of graft obtained in the second procedure was 45 mL (range, 28-65 mL). In the authors' series, no reaming-related complications were observed. The graft volume was the same or increased during the subsequent intramedullary reaming in all but 1 case, suggesting that the intramedullary canal is a potentially renewable source for bone graft. There were no complications related to the sequential reaming procedure. Overall, the authors' data suggest that sequential reaming with the RIA has the potential to safely and effectively provide a large quantity of bone graft on multiple occasions.
Collapse
|
23
|
Isolation of human dermis derived mesenchymal stem cells using explants culture method: expansion and phenotypical characterization. Cell Tissue Bank 2014; 16:209-18. [PMID: 25163610 DOI: 10.1007/s10561-014-9471-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022]
Abstract
Recent studies have reported that stem cells can be isolated from a wide range of tissues including bone marrow, fatty tissue, adipose tissue and placenta. Moreover, several studies also suggest that skin dermis could serve as a source of stem cells, but are of unclear phenotype. Therefore, we isolated and investigated to determine the potential of stem cell within human skin dermis. We isolated cells from human dermis, termed here as human dermis-derived mesenchymal stem cells (hDMSCs) which is able to be isolated by using explants culture method. Our method has an advantage over the enzymatic method as it is easier, less expensive and less cell damage. hDMSCs were maintained in basal culture media and proliferation potential was measured. hDMSCs were highly proliferative and successfully expanded with no additional growth factor. In addition, hDMSCs revealed normal karyotype and expressed high levels of CD90, CD73 and CD105 while did not express the surface markers for CD34, CD45 and HLA-DR. Also, we confirmed that hDMSCs possess the capacity to differentiate into multiple lineage including adipocyte, osteocyte, chondrocyte and precursor of hepatocyte lineage. Considering these results, we suggest that hDMSCs might be a valuable source of stem cells and could potentially be a useful source of clinical application.
Collapse
|
24
|
Blazquez-Martinez A, Chiesa M, Arnalich F, Fernandez-Delgado J, Nistal M, De Miguel MP. c-Kit identifies a subpopulation of mesenchymal stem cells in adipose tissue with higher telomerase expression and differentiation potential. Differentiation 2014; 87:147-60. [PMID: 24713343 DOI: 10.1016/j.diff.2014.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 01/24/2014] [Accepted: 02/24/2014] [Indexed: 12/14/2022]
Abstract
The stromal vascular fraction (SVF) of adipose tissue is an easy to obtain source of adipose tissue-derived stem cells (ADSCs). We and others have achieved significant but suboptimal therapeutic effects with ADSCs in various settings, mainly due to low rates of differentiation into specific cell types and with the downside of undesired side effects as a consequence of the undifferentiated ADSCs. These data prompted us to find new stem cell-specific markers for ADSCs and/or subpopulations with higher differentiation potential to specific lineages. We found a subpopulation of human ADSCs, marked by c-Kit positiveness, resides in a perivascular location, and shows higher proliferative activity and self-renewal capacity, higher telomerase activity and expression, higher in vitro adipogenic efficiency, a higher capacity for the maintenance of cardiac progenitors, and higher pancreatogenic and hepatogenic efficiency independently of CD105 expression. Our data suggests that the isolation of ADSC subpopulations with anti-c-Kit antibodies allows for the selection of a more homogeneous subpopulation with increased cardioprotective properties and increased adipogenic and endodermal differentiation potential, providing a useful tool for specific therapies in regenerative medicine applications.
Collapse
Affiliation(s)
- A Blazquez-Martinez
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain
| | - M Chiesa
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain
| | - F Arnalich
- Department of Internal Medicine, La Paz University Hospital, Madrid, Spain
| | - J Fernandez-Delgado
- Department of Plastic and Reconstructive Surgery, Santa Cristina Hospital, and Centrocim, Madrid, Spain
| | - M Nistal
- Department of Pathology, La Paz University Hospital, Madrid, Spain
| | - M P De Miguel
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain.
| |
Collapse
|
25
|
Samara S, Dailiana Z, Chassanidis C, Koromila T, Papatheodorou L, Malizos KN, Kollia P. Expression profile of osteoprotegerin, RANK and RANKL genes in the femoral head of patients with avascular necrosis. Exp Mol Pathol 2014; 96:9-14. [PMID: 24200492 DOI: 10.1016/j.yexmp.2013.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/22/2013] [Accepted: 10/25/2013] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Femoral head avascular necrosis (AVN) is a recalcitrant disease of the hip that leads to joint destruction. Osteoprotegerin (OPG), Receptor Activator of Nuclear Factor kappa-B (RANK) and RANK ligand (RANKL) regulate the balance between osteoclasts-osteoblasts. The expression of these genes affects the maturation and function of osteoblasts-osteoclasts and bone remodeling. In this study, we investigated the molecular pathways leading to AVN by studying the expression profile of OPG, RANK and RANKL genes. MATERIAL AND METHODS Quantitative Real Time-PCR was performed for evaluation of OPG, RANK and RANKL expression. Analysis was based on parallel evaluation of mRNA and protein levels in normal/necrotic sites of 42 osteonecrotic femoral heads (FHs). OPG and RANKL protein levels were estimated by western blotting. RESULTS The OPG mRNA levels were higher (insignificantly) in the necrotic than the normal site (p > 0.05). Although the expression of RANK and RANKL was significantly lower than OPG in both sites, RANK and RANKL mRNA levels were higher in the necrotic part than the normal (p < 0.05). Protein levels of OPG and RANKL showed no remarkable divergence. CONCLUSIONS Our results indicate that differential expression mechanisms for OPG, RANK and RANKL that could play an important role in the progress of bone remodeling in the necrotic area, disturbing bone homeostasis. This finding may have an effect on the resulting bone destruction and the subsequent collapse of the hip joint.
Collapse
Affiliation(s)
- Stavroula Samara
- Laboratory of Medical Genetics and Cytogenetics, Faculty of Medicine, University of Thessalia, Larissa, Greece
| | - Zoe Dailiana
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Thessalia, Larissa, Greece
| | - Christos Chassanidis
- Laboratory of Medical Genetics and Cytogenetics, Faculty of Medicine, University of Thessalia, Larissa, Greece
| | - Theodora Koromila
- Department of Genetics & Biotechnology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Loukia Papatheodorou
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Thessalia, Larissa, Greece
| | - Konstantinos N Malizos
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Thessalia, Larissa, Greece; Department of Biomedical Research and Technology, Center for Research and Technology, Thessaly (CERETETH), Larissa, Greece
| | - Panagoula Kollia
- Department of Genetics & Biotechnology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
26
|
Samara S, Dailiana Z, Varitimidis S, Chassanidis C, Koromila T, Malizos KN, Kollia P. Bone morphogenetic proteins (BMPs) expression in the femoral heads of patients with avascular necrosis. Mol Biol Rep 2013; 40:4465-4472. [PMID: 23649763 DOI: 10.1007/s11033-013-2538-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/29/2013] [Indexed: 02/07/2023]
Abstract
Avascular necrosis (AVN) is a disorder of the bone repair process which usually results in femoral head (FH) destruction. Bone morphogenetic proteins (BMPs) are the key proteins regulating bone remodelling and healing. BMPs gene expression levels were analyzed in the normal and necrotic sites of osteonecrotic FHs. Quantitative RT-PCR for BMP-2, -4, -6, -7 genes was performed in bone tissue samples from 47 osteonecrotic FHs. Protein levels of BMP-2, -4, -6 were estimated by Western Blot. Statistical analysis was performed using the Wilcoxon signed rank test. BMP-2 and BMP-6 mRNA levels were higher in the normal than the necrotic site (normal/necrotic: 16.8/6.8 and 1.75/1.64, respectively). On the contrary, BMP-4 mRNA levels were higher in the necrotic (0.75) than the normal (0.62), while BMP-7 mRNA levels were extremely low. At the protein level, BMP-2 continued to have a higher expression in the normal region (normal/necrotic: 0.67/0.64). BMP-4 and -6 were detected at higher levels in the necrotic site (normal/necrotic: 0.51/0.61 for BMP-4, 0.51/0.56 for BMP-6), while BMP-7 was not detectable. Different BMP levels between the normal and necrotic site, as well as discrepancies between the gene and protein expression pattern suggest a different regulation mechanism for BMPs between the two regions of FHs. The understanding of the expression pattern and the correlation of BMPs could lead to a more successful use in the prevention and treatment of AVN.
Collapse
Affiliation(s)
- Stavroula Samara
- Laboratory of Medical Genetics and Cytogenetics, Faculty of Medicine, University of Thessalia, Larissa, Greece
| | | | | | | | | | | | | |
Collapse
|
27
|
Knight MN, Hankenson KD. Mesenchymal Stem Cells in Bone Regeneration. Adv Wound Care (New Rochelle) 2013; 2:306-316. [PMID: 24527352 DOI: 10.1089/wound.2012.0420] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Indexed: 01/14/2023] Open
Abstract
SIGNIFICANCE Mesenchymal stem cells (MSCs) play a key role in fracture repair by differentiating to become bone-forming osteoblasts and cartilage-forming chondrocytes. Cartilage then serves as a template for additional bone formation through the process of endochondral ossification. RECENT ADVANCES Endogenous MSCs that contribute to healing are primarily derived from the periosteum, endosteum, and marrow cavity, but also may be contributed from the overlying muscle or through systemic circulation, depending on the type of injury. A variety of growth factor signaling pathways, including BMP, Wnt, and Notch signaling, influence MSC proliferation and differentiation. These MSCs can be therapeutically manipulated to promote differentiation. Furthermore, MSCs can be harvested, cultivated, and delivered to promote bone healing. CRITICAL ISSUES Pharmacologically manipulating the number and differentiation capacity of endogenous MSCs is one potential therapeutic approach to improve healing; however, ideal agents to influence signaling pathways need to be developed and additional therapeutics that activate endogenous MSCs are needed. Whether isolated and purified, MSCs participate directly in the healing process or serve a bystander effect and indirectly influence healing is not well defined. FUTURE DIRECTIONS Studies must focus on better understanding the regulation of endogenous MSCs durings fracture healing. This will reveal novel molecules and pathways to therapeutically target. Similarly, while animal models have demonstrated efficacy in the delivery of MSCs to promote healing, more research is needed to understand ideal donor cells, cultivation methods, and delivery before stem cell therapy approaches can be utilized to repair bone.
Collapse
Affiliation(s)
- M. Noelle Knight
- Veterinary Medical Scientist Training Program, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kurt D. Hankenson
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
28
|
Labusca LS, Botez P, Zugun Eloae F, Mashayekhi K. Stem cells derived from osteoarthritic knee mesenchymal tissues: a pilot study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2013; 23:169-176. [PMID: 23412448 DOI: 10.1007/s00590-012-0949-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/20/2012] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is a promising approach for addressing musculoskeletal disorders. Successful implementation of regenerative therapies is based upon existence of reliable, easy accessible cell sources. Mesenchymal tissues removed during total knee replacement (TKR) were investigated as a potential autologous stem cell source. Materials and methods Samples were collected from patients undergoing primary TKR mononuclear cells from adipose and synovial tissue; subchondral trabecular bone and osteoarthritic cartilage were isolated and assessed in terms of mesenchymal stem cells (MSC) content. Results MSCs obtained from all the investigated tissue types and from all donors showed proliferative, differentiation and surface markers characteristic of stemness. Important number of MSCs could be obtained in the first passage (P0). Mesenchymal tissues removed during TJR can qualitatively and quantitatively function as autologous MSC sources to be considered for regenerative therapies.
Collapse
|
29
|
Dani N, Olivero M, Mareschi K, van Duist MM, Miretti S, Cuvertino S, Patané S, Calogero R, Ferracini R, Scotlandi K, Fagioli F, Di Renzo MF. The MET oncogene transforms human primary bone-derived cells into osteosarcomas by targeting committed osteo-progenitors. J Bone Miner Res 2012; 27:1322-34. [PMID: 22367914 DOI: 10.1002/jbmr.1578] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The MET oncogene is aberrantly overexpressed in human osteosarcomas. We have previously converted primary cultures of human bone-derived cells into osteosarcoma cells by overexpressing MET. To determine whether MET transforms mesenchymal stem cells or committed progenitor cells, here we characterize distinct MET overexpressing osteosarcoma (MET-OS) clones using genome-wide expression profiling, cytometric analysis, and functional assays. All the MET-OS clones consistently display mesenchymal and stemness markers, but not most of the mesenchymal–stem cell-specific markers. Conversely, the MET-OS clones express genes characteristic of early osteoblastic differentiation phases, but not those of late phases. Profiling of mesenchymal stem cells induced to differentiate along osteoblast, adipocyte, and chondrocyte lineages confirms that MET-OS cells are similar to cells at an initial phase of osteoblastic differentiation. Accordingly, MET-OS cells cannot differentiate into adipocytes or chondrocytes, but can partially differentiate into osteogenic-matrix-producing cells. Moreover, in vitro MET-OS cells form self-renewing spheres enriched in cells that can initiate tumors in vivo. MET kinase inhibition abrogates the self-renewal capacity of MET-OS cells and allows them to progress toward osteoblastic differentiation. These data show that MET initiates the transformation of a cell population that has features of osteo-progenitors and suggest that MET regulates self-renewal and lineage differentiation of osteosarcoma cells.
Collapse
Affiliation(s)
- Nadia Dani
- Laboratory of Cancer Genetics, Department of Oncological Sciences University of Torino School of Medicine, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Manferdini C, Gabusi E, Grassi F, Piacentini A, Cattini L, Zini N, Filardo G, Facchini A, Lisignoli G. Evidence of specific characteristics and osteogenic potentiality in bone cells from tibia. J Cell Physiol 2011; 226:2675-82. [DOI: 10.1002/jcp.22618] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
31
|
Pountos I, Giannoudis PV, Jones E, English A, Churchman S, Field S, Ponchel F, Bird H, Emery P, McGonagle D. NSAIDS inhibit in vitro MSC chondrogenesis but not osteogenesis: implications for mechanism of bone formation inhibition in man. J Cell Mol Med 2011; 15:525-34. [PMID: 20070439 PMCID: PMC3922374 DOI: 10.1111/j.1582-4934.2010.01006.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The non-steroidal anti-inflammatory drugs (NSAIDs) are widely used for analgesia but may inhibit bone formation. We investigated whether the reported NSAID effect on bone is related to inhibition of bone marrow mesenchymal stem cell (MSC) proliferation and osteogenic and chondrogenic differentiation and evaluated both cyclooxygenase (COX)-1 and COX-2 specific drugs. The effects of seven COX-1 and COX-2 inhibitors on MSC proliferation and osteogenic and chondrogenic differentiation were tested using Vybrant, sodium 3'-[1-(phenylaminocarbonyl)- 3,4-tetrazolium]-bis (4-methoxy-6-nitro) benzene sulfonic acid hydrate (XTT), functional and quantitative assays of MSC differentiation. The MSC expression of COX-1 and COX-2 and prostaglandin E2 (PGE-2) levels were evaluated serially during lineage differentiation by quantitative PCR and ELISA. None of the NSAIDs at broad range of concentration (range 10(-3) to 100 μg/ml) significantly affected MSC proliferation. Surprisingly, MSC osteogenic differentiation inhibition was not evident. However, NSAIDs affected chondrogenic potential with a reduction in sulphated glycosaminoglycans (sGAG) content by 45% and 55% with diclofenac and ketorolac, respectively (P < 0.05 compared to controls). Parecoxib and meloxicam, more COX-2 specific reagents inhibited sGAG to a lesser degree, 22% and 27% respectively (P < 0.05 compared to controls). Cartilage pellet immunohistochemistry confirmed the above results. Pellet chondrogenesis was associated with increased COX-1 expression levels but not COX-2, and COX-1 specific drugs suppressed MSC PGE-2 more than COX-2 specific inhibitors. These findings suggest that NSAIDs may inhibit bone formation via blockage of MSC chondrogenic differentiation which is an important intermediate phase in normal endochondral bone formation.
Collapse
|
32
|
Multi-potent progenitors in freshly isolated and cultured human mesenchymal stem cells: a comparison between adipose and dermal tissue. Cell Tissue Res 2011; 344:85-95. [DOI: 10.1007/s00441-011-1139-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Accepted: 01/26/2011] [Indexed: 12/20/2022]
|
33
|
Abstract
Excessive intramedullary pressure coincident to surgical procedures requiring entrance and surgical manipulation within the intramedullary canal is a problem that was recognized by Gerhard Küntscher, the godfather of intramedullary nailing. He expressed concern about this phenomenon in his early writings during the 1940's. Although he suggested certain technical methods to moderate the event while doing the surgical procedure he had no solution for absolutely preventing its occurrence. This became more of an issue after he introduced motorized reaming in the mid 1950's to improve the strength of intramedullary fixation. The first to demonstrate that pressure could be avoided during intramedullary surgeries were Lorenzi, Olerud and Dankwardt-Lillieström in the late 1960's. Using a method that employed suction evacuation of intramedullary content prior to reaming, and by introducing irrigation while reaming, they were able to achieve negative pressures during their intramedullary work. They proved that if an IM technique did not inject fat throughout the bone and into the organism there were significant benefits both locally and systemically. With impeccable methodology, they showed fat destroyed the vascularity of the bone and inhibited its revascularization. Systemically, its presence was associated with death and morbidity. K.M. Stürmer, using sheep in studies done in the 1980's, further validated the effectiveness of negative pressure reaming to prevent adverse effects associated with reaming. The attempt to create a device to provide these benefits clinically, however, has been challenging. The group in Muenster did work with a rinsing-suction-reamer (RSR) that showed fat introduction with reaming need not be significantly greater than when using an external fixator. In the US, the effort has focused on developing a reamer that integrated suction and irrigation into its design. This instrument has been given the acronym of RIA (reamer/irrigator/aspirator). The rationale and development of this system is detailed in this paper. Now that the intramedullary canal can be reamed using a negative pressure method this domain, as a unique source of biological material, is being increasingly investigated. The cells and tissue harvested from this space have tremendous therapeutic promise.
Collapse
|
34
|
Henrich D, Seebach C, Sterlepper E, Tauchmann C, Marzi I, Frank J. RIA reamings and hip aspirate: a comparative evaluation of osteoprogenitor and endothelial progenitor cells. Injury 2010; 41 Suppl 2:S62-8. [PMID: 21144931 DOI: 10.1016/s0020-1383(10)70012-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Autologous bone grafting represents the gold standard modality to treat atrophic non-unions by virtue of its osteoinductive and osteoconductive properties. The common harvest site is the iliac crest, but there are major concerns due to limited volume and considerable donor site morbidity. Alternative autologous bone graft can be harvested from the femoral bone cavity using a newly developed 'Reamer Irrigator Aspirator' (RIA). Osseous aspirated particles can be recovered with a filter and used as auto-graft. The purpose of this study was to compare the concentration and differentiation potential of mesenchymal stem cells (MSC) and endothelial progenitor cells (EPC) harvested with the RIA technique or from the iliac crest, respectively. RIA aspirate was collected from 26 patients undergoing intramedullary nailing of femur fractures. Iliac crest aspirate was collected from 38 patients undergoing bone graft transplantation. Concentration of MSC and EPC were assessed by means of the MSC colony assay, EPC culture assay and flowcytometry (CD34, CD133, VEGF-R2), respectively. Osteogenic differentiation of MSC's was measured by von Kossa staining. Patients in both groups did not significantly differ regarding their age, gender or pre-existing health conditions. In comparison to aspirates obtained from iliac crest the RIA aspirates from the femur contained a significantly higher percentage of CD34+ progenitor cells, a significantly higher concentration of MSC and a significantly higher concentration of early EPC. The percentage of late EPC did not differ between both sites. Moreover, the capability of MSC for calcium deposition was significantly enhanced in MSC obtained with RIA. Our results show that RIA aspirate is a rich source for different types of autologous progenitor cells, which can be used to accelerate healing of bone and other musculoskeletal tissues.
Collapse
Affiliation(s)
- Dirk Henrich
- Department of Trauma Surgery, Johann-Wolfgang-Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Effects of combinational adenoviral vector-mediated TGFβ3 transgene and shRNA silencing type I collagen on articular chondrogenesis of synovium-derived mesenchymal stem cells. Biotechnol Bioeng 2010; 106:818-28. [DOI: 10.1002/bit.22733] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Freimark D, Pino-Grace P, Pohl S, Weber C, Wallrapp C, Geigle P, Pörtner R, Czermak P. Use of Encapsulated Stem Cells to Overcome the Bottleneck of Cell Availability for Cell Therapy Approaches. ACTA ACUST UNITED AC 2010; 37:66-73. [PMID: 20737048 DOI: 10.1159/000285777] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 12/29/2009] [Indexed: 11/19/2022]
Abstract
Nowadays cell-based therapy is rarely in clinical practice because of the limited availability of appropriate cells. To apply cells therapeutically, they must not cause any immune response wherefore mainly autologous cells have been used up to now. The amount of vital cells in patients is limited, and under certain circumstances in highly degenerated tissues no vital cells are left. Moreover, the extraction of these cells is connected with additional surgery; also the expansion in vitro is difficult. Other approaches avoid these problems by using allo-or even xenogenic cells. These cells are more stable concerning their therapeutic behavior and can be produced in stock. To prevent an immune response caused by these cells, cell encapsulation (e.g. with alginate) can be performed. Certain studies showed that encapsulated allo- and xenogenic cells achieve promising results in treatment of several diseases. For such cell therapy approaches, stem cells, particularly mesenchymal stem cells, are an interesting cell source. This review deals on the one hand with the use of encapsulated cells, especially stem cells, in cell therapy and on the other hand with bioreactor systems for the expansion and differentiation of mesenchymal stem cells in reproducible and sufficient amounts for potential clinical use.
Collapse
Affiliation(s)
- D Freimark
- Institute of Biopharmaceutical Technology, University of Applied Sciences, Giessen, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Patel S, Waltham M, Wadoodi A, Burnand K, Smith A. The role of endothelial cells and their progenitors in intimal hyperplasia. Ther Adv Cardiovasc Dis 2010; 4:129-41. [DOI: 10.1177/1753944710362903] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Intimal hyperplasia leading to restenosis is the major process that limits the success of cardiovascular intervention. The emergence of vascular progenitor cells and, in particular, endothelial progenitor cells has led to great interest in their potential therapeutic value in preventing intimal hyperplasia. We review the mechanism of intimal hyperplasia and highlight the important attenuating role played by a functional endothelium. The role of endothelial progenitor cells in maintaining endothelial function is reviewed and we describe how reduced progenitor cell number and function and reduced endothelial function lead to an increased risk of intimal hyperplasia. We review other potential sources of endothelial cells, including monocytes, mesenchymal stem cells and tissue resident stem cells. Endothelial progenitor cells have been used in clinical trials to reduce the risk of restenosis with varied success. Progenitor cells have huge therapeutic potential to prevent intimal hyperplasia but a more detailed understanding of vascular progenitor cell biology is necessary before further clinical trials are commenced.
Collapse
Affiliation(s)
- S.D. Patel
- King's College London BHF Centre Cardiovascular Division, NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - M. Waltham
- King's College London BHF Centre Cardiovascular Division, NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - A. Wadoodi
- King's College London BHF Centre Cardiovascular Division, NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - K.G. Burnand
- King's College London BHF Centre Cardiovascular Division, NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - A. Smith
- Academic Department of Surgery, St Thomas' Hospital, Lambeth Palace Road, London SE1 7EH, UK, King's College London BHF Centre, Cardiovascular Division, NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust, London, UK,
| |
Collapse
|
38
|
Yoshii T, Sotome S, Torigoe I, Maehara H, Sugata Y, Yamada T, Shinomiya K, Okawa A. Isolation of Osteogenic Progenitor Cells from Trabecular Bone for Bone Tissue Engineering. Tissue Eng Part A 2010; 16:933-42. [DOI: 10.1089/ten.tea.2009.0105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Toshitaka Yoshii
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Shinichi Sotome
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
- Development Division of Advanced Orthopaedic Therapeutics, Graduate School, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Ichiro Torigoe
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Hidetsugu Maehara
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Yumi Sugata
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Tsuyoshi Yamada
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Kenichi Shinomiya
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
- Global Center of Excellence (GCOE) Program, International Research Center for Molecular Science in Tooth and Bone Disease, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
- Hard Tissue Genome Research Center, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
- Core to Core Program for Advanced Bone and Joint Science, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Atsushi Okawa
- Section of Orthopaedic and Spinal Surgery, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
39
|
A protocol for isolation and culture of mesenchymal stem cells from mouse compact bone. Nat Protoc 2010; 5:550-60. [DOI: 10.1038/nprot.2009.238] [Citation(s) in RCA: 406] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
40
|
Taylor SE, Smith RKW, Clegg PD. Mesenchymal stem cell therapy in equine musculoskeletal disease: scientific fact or clinical fiction? Equine Vet J 2010; 39:172-80. [PMID: 17378447 DOI: 10.2746/042516407x180868] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The goal in the therapeutic use of mesenchymal stem cells (MSCs) in musculoskeletal disease is to harness the regenerative nature of these cells focussing on their potential to grow new tissues and organs to replace damaged or diseased tissue. Laboratory isolation of MSCs is now well established and has recently been demonstrated for equine MSCs. Stem cell science has attracted considerable interest in both the scientific and clinical communities because of its potential to regenerate tissues. Research into the use of MSCs in tissue regeneration in general reflects human medical needs, however, the nature, prevalence and prognosis of superficial digital flexor tendonitis has put equine veterinary science at the forefront of tendon regeneration research. Much has been investigated and learnt but it must be appreciated that in spite of this, the field is still relatively young and both communities must prepare themselves for considerable time and effort to develop the technology into a highly efficient treatments. The promise of functional tissue engineering to replace old parts with new fully justifies the interest. At present, however, it is important to balance the understanding of our current limitations with a desire to progress the technology.
Collapse
Affiliation(s)
- S E Taylor
- Department of Veterinary Clinical Science, University of Liverpool, Leahurst, Chester High Road, Neston, Cheshire CH64 7TE, UK
| | | | | |
Collapse
|
41
|
Qin Y, Ji H, Wu Y, Liu H. Chromosomal instability of murine adipose tissue-derived mesenchymal stem cells in long-term culture and development of cloned embryos. CLONING AND STEM CELLS 2009; 11:445-52. [PMID: 19594392 DOI: 10.1089/clo.2009.0006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mice are the most commonly used laboratory animals for research, and some mouse stem cells, such as induced pluripotent stem cells, embryonic stem cells, and mesenchymal stem cells (MSCs), are also widely used in basic research. It is thus important to know if these stem cells maintain their genomic stability when cultured. Murine bone marrow-derived mesenchymal stem cells (BMSCs) appear to undergo spontaneous transformation in vitro. Murine adipose tissue-derived mesenchymal stem cells (ADSCs), like BMSCs, have the potential to differentiate into multiple lineages. In this study, we used G-banding, induction of multiple-lineage differentiation, flow cytometry, and nuclear transfer (NT), and found that murine ADSCs also displayed chromosomal instability in long-term culture. Furthermore, we performed NT using murine ADSCs to study the nuclear reprogramming ability of undifferentiated adult stem cells and to find a new efficient donor for NT. Using the stem cells did not increase the percentage of NT embryos that developed to the morula/blastocyst stage, compared with cloned embryos from cumulus cells. This may be because the stem cells displayed chromosomal instability. This is the first reported study of the use of ADSCs for NT in mice. ADSCs could provide an alternative donor cell type for NT in other species, with the advantages of easy harvesting involving little or no pain or trauma.
Collapse
Affiliation(s)
- Yiren Qin
- Department of Histology and Embryology, Harbin Medical University, Harbin, PR China
| | | | | | | |
Collapse
|
42
|
Coipeau P, Rosset P, Langonne A, Gaillard J, Delorme B, Rico A, Domenech J, Charbord P, Sensebe L. Impaired differentiation potential of human trabecular bone mesenchymal stromal cells from elderly patients. Cytotherapy 2009; 11:584-94. [PMID: 19626496 DOI: 10.1080/14653240903079385] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS Advances in bone tissue engineering with mesenchymal stromal cells (MSC) as an alternative to conventional orthopedic procedures has opened new horizons for the treatment of large bone defects. Bone marrow (BM) and trabecular bone are both sources of MSC. Regarding clinical use, we tested the potency of MSC from different sources. METHODS We obtained MSC from 17 donors (mean age 64.6 years) by extensive washing of trabecular bone from the femoral head and trochanter, as well as BM aspirates of the iliac crest and trochanter. The starting material was evaluated by histologic analysis and assessment of colony-forming unit-fibroblasts (CFU-F). The MSC populations were compared for proliferation and differentiation potential, at RNA and morphologic levels. RESULTS MSC proliferation potential and immunophenotype (expression of CD49a, CD73, CD90, CD105, CD146 and Stro-1) were similar whatever the starting material. However, the differentiation potential of MSC obtained by bone washing was impaired compared with aspiration; culture-amplified cells showed few Oil Red O-positive adipocytes and few mineralized areas and formed inconsistent Alcian blue-positive high-density micropellets after growth under adipogenic, osteogenic and chondrogenic conditions, respectively. MSC cultured with 1 ng/mL fibroblast growth factor 2 (FGF-2) showed better differentiation potential. CONCLUSIONS Trabecular bone MSC from elderly patients is not good starting material for use in cell therapy for bone repair and regeneration, unless cultured in the presence of FGF-2.
Collapse
Affiliation(s)
- Patrick Coipeau
- Department of Orthopedic Surgery, University Hospital, Tours, France
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhang ZM, Jiang LS, Jiang SD, Dai LY. Osteogenic potential and responsiveness to leptin of mesenchymal stem cells between postmenopausal women with osteoarthritis and osteoporosis. J Orthop Res 2009; 27:1067-73. [PMID: 19215023 DOI: 10.1002/jor.20846] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The aim of this study was to compare the osteogenic potential and responsiveness to leptin of mesenchymal stem cells (MSCs) from bone marrow between postmenopausal women with osteoarthritis (OA) and osteoporosis (OP). MSCs of the proximal femur from OA and OP donors were cultured under control and different experimental mediums. After verifying the availability of primary cells, their osteogenic potential and responsiveness to leptin were compared between two groups. Similar patterns of cell growth were shown in both OA and OP groups. However, after the sixth passage, the viability of undifferentiated cells decreased more in OP than in OA donors. Under the same osteogenic supplements condition, the mRNA expression of osteogenesis-specific genes, osteocalcin (OC) and alkaline phosphatase (ALP) were higher in OA group. Comparison of bone matrix mineralization was parallel to that of mRNA expression. The level of bone-specific ALP (BAP) was higher in cells from donors with OA, whereas osteoprotegerin (OPG) was higher in OP group. This difference in BAP expression proved to be insignificant after the administration of leptin. Although leptin upregulated the expression of OPG, a significant difference still existed between OA and OP. In conclusion, differential osteogenic potential and responsiveness to leptin of MSCs were noted between postmenopausal women with OA and OP. Differential biological behavior of MSCs seems to be partly related to the different distribution of bone mass between OA and OP populations.
Collapse
Affiliation(s)
- Zi-Ming Zhang
- Department of Orthopaedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | | | | | | |
Collapse
|
44
|
Freimark D, Czermak P. Cell-based regeneration of intervertebral disc defects: review and concepts. Int J Artif Organs 2009; 32:197-203. [PMID: 19569027 DOI: 10.1177/039139880903200403] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
During the last century low back pain has emerged as a widespread disease often caused by intervertebral disc degeneration (IDD). IDD is a complex problem in which a variety of causes play a role. As IDD causes high costs, corporate interest has led to a number of therapies being developed. Today, these therapies focus not only on minimizing the pain caused by this disease but also on restoring intervertebral disc function. These approaches are often biological and aim to stimulate the regeneration of the intervertebral disc by injection of activator proteins, biomaterials, different cell types or complex cell matrix composites. Genetic engineering of disc cells and in vitro tissue engineering also offer a possibility for curing IDD. This article gives an overview of these concepts.
Collapse
Affiliation(s)
- Denise Freimark
- Institute of Biopharmaceutical Technology, University of Applied Sciences Giessen-Friedberg, Giessen, Germany.
| | | |
Collapse
|
45
|
Sanchez-Guijo FM, Blanco JF, Cruz G, Muntion S, Gomez M, Carrancio S, Lopez-Villar O, Barbado MV, Sanchez-Abarca LI, Blanco B, Briñon JG, del Cañizo MC. Multiparametric comparison of mesenchymal stromal cells obtained from trabecular bone by using a novel isolation method with those obtained by iliac crest aspiration from the same subjects. Cell Tissue Res 2009; 336:501-7. [DOI: 10.1007/s00441-009-0778-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 02/04/2009] [Indexed: 01/12/2023]
|
46
|
Abstract
This review article describes bone remodeling in the context of osteonecrosis as a bone disease, the use of stem cells in bone and vascular diseases, and cellular therapy in osteonecrosis.
Collapse
Affiliation(s)
- Valérie Gangji
- Department of Rheumatology and Physical Medicine, Erasme University Hospital, Université Libre de Bruxelles, 808 Route de Lennik, Bruxelles 1070, Belgium.
| | | |
Collapse
|
47
|
Biological characterization of long-term cultured human mesenchymal stem cells. Arch Pharm Res 2009; 32:117-26. [PMID: 19183884 DOI: 10.1007/s12272-009-1125-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 12/17/2008] [Accepted: 12/26/2008] [Indexed: 01/02/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have generated a great deal of interest in clinical applications. The reason is that they may have the plasticity needed to differentiate into multiple lineages and the ability to expand ex vivo. For the therapeutic applications of hMSCs to be of practical use, it is crucial to assess the efficacy and safety of hMSCs in long-term ex vivo expansion. In this study, we cultured hMSCs by population doubling (PD) 60, and investigated their growth, osteogenic and adipogenic differential abilities, change of surface markers, telomerase activity, telomere length, and gene expression related to tumorigenesis. An in vivo tumorigenesis assay was also carried out. In long-term expanded hMSCs, the cells became aged above PD 30 and their adipogenic and osteogenic differentiation potential decreased. Telomerase activity unchanged whereas telomere length decreased and karyotypes were not changed. Gene expressions related to tumorigenesis decreased in proportion as the PD of hMSCs increased. In vivo transplantation of long-term cultured hMSCs to nude mice did not result in tumor formation. These findings suggest that diverse tests for cellular therapy should be considered during the ex vivo culture of hMSCs, particularly when a prolonged and extended propagation period is required.
Collapse
|
48
|
Mabuchi Y, Morikawa S, Suzuki S, Sunabori T, Okanoa H, Matsuzaki Y. Prospective isolation and identification of human mesenchymal stem cells by flow cytometry. Inflamm Regen 2009. [DOI: 10.2492/inflammregen.29.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
49
|
Trojani C, Balaguer T, Boukhechba F, Carle GF, Boileau P, Rochet N. Inventaire des stratégies cellulaires en ingénierie tissulaire de reconstruction osseuse. ACTA ACUST UNITED AC 2008; 94:1-11. [DOI: 10.1016/j.rco.2007.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2007] [Indexed: 01/09/2023]
|
50
|
A simple, lanthanide-based method to enhance the transduction efficiency of adenovirus vectors. Gene Ther 2008; 15:357-63. [PMID: 18283289 DOI: 10.1038/sj.gt.3303092] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based upon the powerful bridging and charge-masking properties of lanthanide cations (Ln3+), we have investigated their use to improve the transduction efficiency of adenovirus vectors. Using a luciferase marker gene, it was possible to increase transgene expression by the murine mesenchymal stem cell line C3H10T(1/2) by up to four log orders when using very low multiplicities of infection in conjunction with Ln3+; La3+ was superior to Gd3+, Y3+ and Lu3+ in this regard. All Ln3+ were more effective than Ca2+. Flow cytometry, using a green fluorescent protein marker gene, confirmed that La3+ increased both the percentage of transduced cells and the level of transgene expression per cell. Transduction of primary cultures of a variety of different mesenchymal cells from human, rabbit, bovine and rat sources, as well as gene transfer to synovium and muscle in vivo, was also greatly enhanced. Our findings suggest that this lanthanide-based method holds much promise for expediting both experimental and clinical applications of gene transfer with adenoviral vectors.
Collapse
|