1
|
Hayashi Y, Kawabata KC, Tanaka Y, Uehara Y, Mabuchi Y, Murakami K, Nishiyama A, Kiryu S, Yoshioka Y, Ota Y, Sugiyama T, Mikami K, Tamura M, Fukushima T, Asada S, Takeda R, Kunisaki Y, Fukuyama T, Yokoyama K, Uchida T, Hagihara M, Ohno N, Usuki K, Tojo A, Katayama Y, Goyama S, Arai F, Tamura T, Nagasawa T, Ochiya T, Inoue D, Kitamura T. MDS cells impair osteolineage differentiation of MSCs via extracellular vesicles to suppress normal hematopoiesis. Cell Rep 2022; 39:110805. [PMID: 35545056 DOI: 10.1016/j.celrep.2022.110805] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/15/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells (HSCs), characterized by ineffective hematopoiesis and frequent progression to leukemia. It has long remained unresolved how MDS cells, which are less proliferative, inhibit normal hematopoiesis and eventually dominate the bone marrow space. Despite several studies implicating mesenchymal stromal or stem cells (MSCs), a principal component of the HSC niche, in the inhibition of normal hematopoiesis, the molecular mechanisms underlying this process remain unclear. Here, we demonstrate that both human and mouse MDS cells perturb bone metabolism by suppressing the osteolineage differentiation of MSCs, which impairs the ability of MSCs to support normal HSCs. Enforced MSC differentiation rescues the suppressed normal hematopoiesis in both in vivo and in vitro MDS models. Intriguingly, the suppression effect is reversible and mediated by extracellular vesicles (EVs) derived from MDS cells. These findings shed light on the novel MDS EV-MSC axis in ineffective hematopoiesis.
Collapse
Affiliation(s)
- Yasutaka Hayashi
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Minatojimaminami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Kimihito C Kawabata
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Division of Hematology/Medical Oncology, Department of Medicine, Weill-Cornell Medical College, Cornell University, NY 10021, USA
| | - Yosuke Tanaka
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yasufumi Uehara
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Koichi Murakami
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0043, Japan; Advanced Medical Research Center, Yokohama City University, Yokohama 236-0043, Japan
| | - Akira Nishiyama
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0043, Japan
| | - Shigeru Kiryu
- Department of Radiology, International University of Health and Welfare Narita Hospital, Chiba 286-8686, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yasunori Ota
- Department of Pathology, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuki Sugiyama
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Keiko Mikami
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Moe Tamura
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
| | - Tsuyoshi Fukushima
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Shuhei Asada
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Reina Takeda
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yuya Kunisaki
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Tomofusa Fukuyama
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tomoyuki Uchida
- Department of Hematology, Eiju General Hospital, Tokyo 110-8645, Japan
| | - Masao Hagihara
- Department of Hematology, Eiju General Hospital, Tokyo 110-8645, Japan
| | - Nobuhiro Ohno
- Department of Hematology, Kanto Rosai Hospital, Kawasaki 211-8510, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo 141-8625, Japan
| | - Arinobu Tojo
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | | | - Susumu Goyama
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
| | - Fumio Arai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomohiko Tamura
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0043, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Minatojimaminami-machi, Chuo-ku, Kobe 650-0047, Japan.
| | - Toshio Kitamura
- Division of Cellular Therapy, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
2
|
Teufelsbauer M, Lang C, Plangger A, Rath B, Moser D, Staud C, Radtke C, Neumayer C, Hamilton G. Effects of metformin on human bone-derived mesenchymal stromal cell-breast cancer cell line interactions. Med Oncol 2022; 39:54. [PMID: 35150338 PMCID: PMC8840908 DOI: 10.1007/s12032-022-01655-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
Metformin is used to treat patients with type 2 diabetes mellitus and was found to lower the incidence of cancer. Bone metastasis is a common impairment associated with advanced breast cancer. The present study investigated the effects of metformin on human bone-derived mesenchymal stromal cells (BM-MSC)—breast cancer cell line interactions. BM-MSCs grown from box chisels were tested for growth-stimulating and migration-controlling activity on four breast cancer cell lines either untreated or after pretreatment with metformin. Growth stimulation was tested in MTT tests and migration in scratch assays. Furthermore, the expression of adipokines of BM-MSCs in response to metformin was assessed using Western blot arrays. Compared to breast cancer cell lines (3.6 ± 1.4% reduction of proliferation), 500 µM metformin significantly inhibited the proliferation of BM-MSC lines (mean 12.3 ± 2.2 reduction). Pretreatment of BM-MSCs with metformin showed variable effects of the resulting conditioned media (CM) on breast cancer cell lines depending on the specific BM-MSC—cancer line combination. Metformin significantly reduced the migration of breast cancer cell lines MDA-MB-231 and MDA-MB-436 in response to CM of drug-pretreated BM-MSCs. Assessment of metformin-induced alterations in the expression of adipokines by BM-MSC CM indicated increased osteogenic signaling and possibly impairment of metastasis. In conclusion, the anticancer activities of metformin are the result of a range of direct and indirect mechanisms that lower tumor proliferation and progression. A lower metformin-induced protumor activity of BM-MSCs in the bone microenvironment seem to contribute to the positive effects of the drug in selected breast cancer patients.
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Clemens Lang
- Department of Trauma Surgery, Sozialmedizinisches Zentrum Ost, Donauspital, Vienna, Austria
| | - Adelina Plangger
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Doris Moser
- Department of Cranio, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Pengjam Y, Syazwani N, Inchai J, Numit A, Yodthong T, Pitakpornpreecha T, Panichayupakaranant P. High water-soluble curcuminoids-rich extract regulates osteogenic differentiation of MC3T3-E1 cells: Involvement of Wnt/β-catenin and BMP signaling pathway. CHINESE HERBAL MEDICINES 2021; 13:534-540. [PMID: 36119369 PMCID: PMC9476497 DOI: 10.1016/j.chmed.2021.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The present study aimed to evaluate the effect of a high water-soluble curcuminoids-rich extract (CRE) in a solid dispersion form (CRE-SD) using polyvinylpyrrolidone K30 on osteogenic induction of MC3T3-E1 cells. METHODS CRE was pre-purified using a microwave assisted extraction couple with a Diaion® HP-20 column chromatography. The osteoblastic cell proliferation and differentiation potentials of CRE-SD in MC3T3-E1 cells were tested by cell viability, alkaline phosphatase (ALP) activity, and Alizarin red S activity assays. The mRNA expressions of osteoblast-specific genes and underline mechanisms were assessed by a real time PCR and western blot analysis. RESULTS CRE-SD 50 µg/mL increased alkaline phosphatase (ALP) activity, an early differentiation marker of osteoblasts in both MC3T3-E1 cells and non-osteogenic mouse pluripotent cell line, C3H10T1/2, indicating the action of CRE-SD was not cell-type specific. Alizarin red S activity showed a significant amount of calcium deposition in cells treated with CRE-SD. CRE-SD also upregulated the mRNA expression levels of transcription factors that favor osteoblast differentiation including Bmp-2, Runx2 and Collagen 1a, in a dose dependent manner. Western blot analysis revealed that noggin attenuated CRE-SD-promoted expressions of Bmp-2 and Runx2 proteins. siRNA mediated blocking of Wnt/β-catenin signaling pathway also annulled the influence of CRE-SD, indicating Wnt/β-catenin dependent activity. Inhibition of the different signaling pathways abolished the influence of CRE-SD on ALP activity, confirming that CRE-SD induced MC3T3-E1 cells into osteoblasts through Wnt/β-catenin and BMP signaling pathway. CONCLUSION These results collectively demonstrate that CRE-SD may be a potential therapeutic agent for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yutthana Pengjam
- Faculty of Medical Technology, Prince of Songkla University, Hat-Yai 90110, Thailand
| | - Nurul Syazwani
- Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai 90112, Thailand
| | - Jakkapong Inchai
- Faculty of Medical Technology, Prince of Songkla University, Hat-Yai 90110, Thailand
| | - Amornkan Numit
- Faculty of Medical Technology, Prince of Songkla University, Hat-Yai 90110, Thailand
| | | | | | | |
Collapse
|
4
|
Khorsand B, Elangovan S, Hong L, Kormann MSD, Salem AK. A bioactive collagen membrane that enhances bone regeneration. J Biomed Mater Res B Appl Biomater 2019; 107:1824-1832. [PMID: 30466196 PMCID: PMC6531367 DOI: 10.1002/jbm.b.34275] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/08/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Membranes are an integral component of guided bone regeneration protocols. This pre-clinical study was aimed at enhancing the bioactivity of collagen membranes by incorporating plasmid DNA (pDNA) or chemically modified RNA (cmRNA) encoding bone morphogenetic protein-9 (BMP-9). In addition, we also endeavored to harness the regenerative potential of the periosteum by creating perforations in the membrane. Nanoplexes of polyethylenimine (PEI)-nucleic acids (PEI-pDNA or PEI-cmRNA encoding BMP-9) were incorporated into commercially obtained and perforated collagen membranes (PCM) to produce PCM-pDNA(BMP-9) or PCM-cmRNA(BMP-9). After structural characterization, the biodegradation kinetics of PCM, PCM-pDNA(BMP-9) and PCM-cmRNA(BMP-9) were assessed in simulated body fluid in vitro. Using a 24-well transwell plate system with bone marrow stromal cells (BMSCs) in the lower chamber and the PCM to be tested in the upper chamber, the in vitro bioactivity of different PCMs was evaluated by measuring various markers for osteogenesis in BMSCs. Alkaline phosphatase activity was assessed in BMSCs, after 7 and 11 days of exposure to PCM, PCM-pDNA(BMP-9), or PCM-cmRNA(BMP-9). Similarly, calcium deposition and Alizarin red staining in BMSCs were assessed after 14 days of exposure to the three different types of PCM. PCMs were then tested in vivo using the calvarial defect model in rats. After 4 weeks, animals were euthanized and bone specimens were harvested for micro-computed tomography and histological assessments. Incorporation of pDNA or cmRNA did not alter the biodegradation profile of PCMs. Alkaline phosphatase activity trended toward being higher in BMSCs exposed to PCM-cmRNA(BMP-9) or PCM-pDNA(BMP-9), when compared to BMSCs alone. Similar trends were observed when calcium deposition and alizarin red staining was evaluated. Calvarial bone defects treated with PCM-cmRNA(BMP-9) resulted in significantly higher bone volume/total volume % (BV/TV%), when compared to empty defects and trended toward being higher than defects treated with PCM-pDNA(BMP-9) and PCM alone. We demonstrate for the first time that resorbable PCM can be utilized to efficiently deliver pDNA and cmRNA of interest. The released pDNA and cmRNA encoding BMP-9 in this assessment was shown to be functional in vitro as well as in vivo. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1824-1832, 2019.
Collapse
Affiliation(s)
- Behnoush Khorsand
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa
| | - Satheesh Elangovan
- Department of Periodontics, University of Iowa College of Dentistry, Iowa City, Iowa
| | - Liu Hong
- Department of Prosthodontics University of Iowa College of Dentistry, Iowa City, Iowa
| | - Michael S D Kormann
- Department of Translational Genomics and Gene Therapy, University of Tübingen, Wilhelmstr. 56, Tübingen, Germany
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa
- Department of Periodontics, University of Iowa College of Dentistry, Iowa City, Iowa
| |
Collapse
|
5
|
Besikcioglu HE, Sarıbas GS, Ozogul C, Tiryaki M, Kilic S, Pınarlı FA, Gulbahar O. Determination of the effects of bone marrow derived mesenchymal stem cells and ovarian stromal stem cells on follicular maturation in cyclophosphamide induced ovarian failure in rats. Taiwan J Obstet Gynecol 2019; 58:53-59. [PMID: 30638481 DOI: 10.1016/j.tjog.2018.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2018] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Chemotherapy causes depletion of primordial follicles that leads to premature ovarian failure in female cancer survivals. We investigated the effect of bone marrow derived mesenchymal (BMMSCs) and ovarian stromal stem cells (OSSCs) on follicle maturation in chemotherapy induced ovarian failure. MATERIAL AND METHODS Thirty six Wistar Albino female rats were divided into three groups. Cyclophosphamide at a dose of 200 mg/kg was intraperitoneally (IP) given to the rats in all groups two times. 4 × 106 BMMSCs (IP) was injected to the group-2 and 4 × 106 OSSCs (IP) was injected to the group-3. Serum Anti-Müllerian Hormone (AMH) levels was determined with ELISA and primordial follicles were counted for investigation of primordial follicle reserve. The ovarian structure were evaluated histomorphologically. Localization of BrdU labeled stem cells, the expression of the cell cycle regulator p34Cdc2, gap junction protein p-connexin43 and intraovarian regulators of folliculogenesis Bone Morphogenic Protein 6 and 15 (BMP-6 and BMP-15) were investigated by immunohistochemistry. RESULTS The immunstaining of BMP-6 was higher in oocytes of group-3 more than group-1 and group-2. The immunpositivity of p34cdc2 and BMP-15 were also higher in follicular cells of group-3 than the other groups. The presence of p-connexin43 in group-3 was determined more than group-1 and group-2. The ovarian follicles with normal histological structure were observed just in group-3. Although, The AMH levels were decreased in rats from all groups at the end of experimental procedure the primordial follicle counts in group-3 was significantly higher than group-1. CONCLUSION Our findings suggest that OSSCs have more protective effect on follicle maturation than BMMSCs in cyclophosphamide induced ovarian damage.
Collapse
Affiliation(s)
| | - Gulistan Sanem Sarıbas
- Department of Histology and Embryology, Faculty of Medicine, Kırşehir Ahi Evran University, Kırşehir, Turkey
| | - Candan Ozogul
- Department of Histology and Embryology, Faculty of Medicine Gazi University, Ankara, Turkey.
| | - Meral Tiryaki
- Department of Pathology, Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Sevtap Kilic
- Department of Obstetrics and Gynecology, Faculty of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Ferda Alpaslan Pınarlı
- Genetic Diagnostic Center, Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
6
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-β family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| | - Vicki Rosen
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
7
|
Huang B, Wu Z, Ding S, Yuan Y, Liu C. Localization and promotion of recombinant human bone morphogenetic protein-2 bioactivity on extracellular matrix mimetic chondroitin sulfate-functionalized calcium phosphate cement scaffolds. Acta Biomater 2018; 71:184-199. [PMID: 29355717 DOI: 10.1016/j.actbio.2018.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/28/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
Abstract
Localization of recombinant human bone morphogenetic protein-2 (rhBMP-2) with continuous and effective osteogenic stimulation is still a great challenge in the field of bone regeneration. To achieve this aim, rhBMP-2 was tethered on chondroitin sulfate (CS)-functionalized calcium phosphate cement (CPC) scaffolds through specific noncovalent interactions. CS, one of the core glycosaminoglycans, was covalently conjugated onto CPC scaffolds with the assistance of polydopamine (PDA) and further immobilized rhBMP-2 in a biomimetic form. The CPC-PDA-CS scaffolds not only controlled the release kinetics and presentation state of rhBMP-2 but also effectively increased the expression levels of bone morphogenetic protein receptors (BMPRs) and enhanced the recognitions of the remaining rhBMP-2 to BMPRs. Strikingly, the rhBMP-2-loaded CPC-PDA-CS significantly promoted the cellular surface translocation of BMPRs (especially BMPR-IA). In vivo studies demonstrated that, compared with the rhBMP-2 upon CPC and CPC-PDA, the rhBMP-2 upon CPC-PDA-CS exhibited sustained release and induced high quality and more ectopic bone formation. Collectively, these results suggest that rhBMP-2 can be localized within CS-functionalized CPC scaffolds and exert continuous, long-term, and effective osteogenic stimulation. Thus, this work could provide new avenues in mimicking bone extracellular matrix microenvironment and localizing growth factor activity for enhanced bone regeneration. STATEMENT OF SIGNIFICANCE A bioinspired chondroitin sulfate (CS)-functionalized calcium phosphate cement (CPC) platform was developed to tether recombinant human bone morphogenetic protein-2 (rhBMP-2), which could exhibit continuous, long-term, and effective osteogenic stimulation in bone tissue engineering. Compared with rhBMP-2-loaded CPC, the rhBMP-2-loaded CPC-polydopamine-CS scaffolds induced higher expression of bone morphogenetic protein receptors (BMPRs), greater cellular surface translocation of bone morphogenetic protein receptor-IA, higher binding affinity of BMPRs/rhBMP-2, and thus higher activation of the drosophila gene mothers against decapentaplegic protein-1/5/8 (Smad1/5/8) and extracellular-regulated protein kinases-1/2 (ERK1/2) signaling. This work can provide new guidelines for the design of BMP-2-based bioactive materials for bone regeneration.
Collapse
|
8
|
Bearden RN, Huggins SS, Cummings KJ, Smith R, Gregory CA, Saunders WB. In-vitro characterization of canine multipotent stromal cells isolated from synovium, bone marrow, and adipose tissue: a donor-matched comparative study. Stem Cell Res Ther 2017; 8:218. [PMID: 28974260 PMCID: PMC5627404 DOI: 10.1186/s13287-017-0639-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Background The dog represents an excellent large animal model for translational cell-based studies. Importantly, the properties of canine multipotent stromal cells (cMSCs) and the ideal tissue source for specific translational studies have yet to be established. The aim of this study was to characterize cMSCs derived from synovium, bone marrow, and adipose tissue using a donor-matched study design and a comprehensive series of in-vitro characterization, differentiation, and immunomodulation assays. Methods Canine MSCs were isolated from five dogs with cranial cruciate ligament rupture. All 15 cMSC preparations were evaluated using colony forming unit (CFU) assays, flow cytometry analysis, RT-PCR for pluripotency-associated genes, proliferation assays, trilineage differentiation assays, and immunomodulation assays. Data were reported as mean ± standard deviation and compared using repeated-measures analysis of variance and Tukey post-hoc test. Significance was established at p < 0.05. Results All tissue samples produced plastic adherent, spindle-shaped preparations of cMSCs. Cells were negative for CD34, CD45, and STRO-1 and positive for CD9, CD44, and CD90, whereas the degree to which cells were positive for CD105 was variable depending on tissue of origin. Cells were positive for the pluripotency-associated genes NANOG, OCT4, and SOX2. Accounting for donor and tissue sources, there were significant differences in CFU potential, rate of proliferation, trilineage differentiation, and immunomodulatory response. Synovium and marrow cMSCs exhibited superior early osteogenic activity, but when assessing late-stage osteogenesis no significant differences were detected. Interestingly, bone morphogenic protein-2 (BMP-2) supplementation was necessary for early-stage and late-stage osteogenic differentiation, a finding consistent with other canine studies. Additionally, synovium and adipose cMSCs proliferated more rapidly, displayed higher CFU potential, and formed larger aggregates in chondrogenic assays, although proteoglycan and collagen type II staining were subjectively decreased in adipose pellets as compared to synovial and marrow pellets. Lastly, cMSCs derived from all three tissue sources modulated murine macrophage TNF-α and IL-6 levels in a lipopolysaccharide-stimulated coculture assay. Conclusions While cMSCs from synovium, marrow, and adipose tissue share a number of similarities, important differences in proliferation and trilineage differentiation exist and should be considered when selecting cMSCs for translational studies. These results and associated methods will prove useful for future translational studies involving the canine model. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0639-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert N Bearden
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shannon S Huggins
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Kevin J Cummings
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Roger Smith
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, College of Medicine, Texas A&M University, College Station, TX, USA
| | - William B Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
9
|
Khorsand B, Nicholson N, Do AV, Femino JE, Martin JA, Petersen E, Guetschow B, Fredericks DC, Salem AK. Regeneration of bone using nanoplex delivery of FGF-2 and BMP-2 genes in diaphyseal long bone radial defects in a diabetic rabbit model. J Control Release 2017; 248:53-59. [PMID: 28069556 PMCID: PMC5305420 DOI: 10.1016/j.jconrel.2017.01.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/07/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022]
Abstract
Bone fracture healing impairment related to systemic diseases such as diabetes can be addressed by growth factor augmentation. We previously reported that growth factors such as fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) work synergistically to encourage osteogenesis in vitro. In this report, we investigated if BMP-2 and FGF-2 together can synergistically promote bone repair in a leporine model of diabetes mellitus, a condition that is known to be detrimental to union. We utilized two kinds of plasmid DNA encoding either BMP-2 or FGF-2 formulated into polyethylenimine (PEI) complexes. The fabricated nanoplexes were assessed for their size, charge, in vitro cytotoxicity, and capacity to transfect human bone marrow stromal cells (BMSCs). Using diaphyseal long bone radial defects in a diabetic rabbit model it was demonstrated that co-delivery of PEI-(pBMP-2+pFGF-2) embedded in collagen scaffolds resulted in a significant improvement in bone regeneration compared to PEI-pBMP-2 embedded in collagen scaffolds alone. This study demonstrated that scaffolds loaded with PEI-(pBMP-2+pFGF-2) could be an effective way of promoting bone regeneration in patients with diabetes.
Collapse
Affiliation(s)
- Behnoush Khorsand
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, IA, United States
| | - Nate Nicholson
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Anh-Vu Do
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, IA, United States
| | - John E Femino
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - James A Martin
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Emily Petersen
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Brian Guetschow
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Douglas C Fredericks
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, IA, United States.
| |
Collapse
|
10
|
Khorsand B, Elangovan S, Hong L, Dewerth A, Kormann MSD, Salem AK. A Comparative Study of the Bone Regenerative Effect of Chemically Modified RNA Encoding BMP-2 or BMP-9. AAPS JOURNAL 2017; 19:438-446. [PMID: 28074350 DOI: 10.1208/s12248-016-0034-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/20/2016] [Indexed: 02/02/2023]
Abstract
Employing cost-effective biomaterials to deliver chemically modified ribonucleic acid (cmRNA) in a controlled manner addresses the high cost, safety concerns, and lower transfection efficiency that exist with protein and gene therapeutic approaches. By eliminating the need for nuclear entry, cmRNA therapeutics can potentially overcome the lower transfection efficiencies associated with non-viral gene delivery systems. Here, we investigated the osteogenic potential of cmRNA-encoding BMP-9, in comparison to cmRNA-encoding BMP-2. Polyethylenimine (PEI) was used as a vector to increase in vitro transfection efficacy. Complexes of PEI-cmRNA (encoding BMP-2 or BMP-9) were fabricated at an amine (N) to phosphate (P) ratio of 10 and characterized for transfection efficacy in vitro using human bone marrow stromal cells (BMSCs). The osteogenic potential of BMSCs treated with these complexes was determined by evaluating the expression of bone-specific genes as well as through the detection of bone matrix deposition. It was found that alkaline phosphatase (ALP) expression 3 days post transfection in the group treated with BMP-9-cmRNA was significantly higher than that in the group that received BMP-2-cmRNA treatment. Alizarin red staining and atomic absorption spectroscopy demonstrated enhanced osteogenic differentiation as evidenced by increased bone matrix production by the BMSCs treated with BMP-9-cmRNA when compared to cells treated with BMP-2-cmRNA. In vivo studies showed increased bone formation in calvarial defects treated with the BMP-9-cmRNA and BMP-2-cmRNA collagen scaffolds when compared to empty defects. The connectivity density of the regenerated bone was higher (2-fold-higher) in the group that received BMP-9-cmRNA compared to BMP-2-cmRNA. Together, these findings suggest that cmRNA-activated matrix encoding osteogenic molecules can provide a powerful strategy for bone regeneration with significant clinical translational potential.
Collapse
Affiliation(s)
- Behnoush Khorsand
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Satheesh Elangovan
- Department of Periodontics, University of Iowa College of Dentistry, Iowa City, Iowa, USA.
| | - Liu Hong
- Department of Prosthodontics, University of Iowa College of Dentistry, Iowa City, Iowa, USA
| | - Alexander Dewerth
- Department of Pediatrics (Section I), Translational Genomics and Gene Therapy, University of Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Michael S D Kormann
- Department of Pediatrics (Section I), Translational Genomics and Gene Therapy, University of Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA. .,Department of Periodontics, University of Iowa College of Dentistry, Iowa City, Iowa, USA.
| |
Collapse
|
11
|
Lee PT, Li WJ. Chondrogenesis of Embryonic Stem Cell-Derived Mesenchymal Stem Cells Induced by TGFβ1 and BMP7 Through Increased TGFβ Receptor Expression and Endogenous TGFβ1 Production. J Cell Biochem 2016; 118:172-181. [PMID: 27292615 DOI: 10.1002/jcb.25623] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
For decades stem cells have proven to be invaluable to the study of tissue development. More recently, mesenchymal stem cells (MSCs) derived from embryonic stem cells (ESCs) (ESC-MSCs) have emerged as a cell source with great potential for the future of biomedical research due to their enhanced proliferative capability compared to adult tissue-derived MSCs and effectiveness of musculoskeletal lineage-specific cell differentiation compared to ESCs. We have previously compared the properties and differentiation potential of ESC-MSCs to bone marrow-derived MSCs. In this study, we evaluated the potential of TGFβ1 and BMP7 to induce chondrogenic differentiation of ESC-MSCs compared to that of TGFβ1 alone and further investigated the cellular phenotype and intracellular signaling in response to these induction conditions. Our results showed that the expression of cartilage-associated markers in ESC-MSCs induced by the TGFβ1 and BMP7 combination was increased compared to induction with TGFβ1 alone. The TGFβ1 and BMP7 combination upregulated the expression of TGFβ receptor and the production of endogenous TGFβs compared to TGFβ1 induction. The growth factor combination also increasingly activated both of the TGF and BMP signaling pathways, and inhibition of the signaling pathways led to reduced chondrogenesis of ESC-MSCs. Our findings suggest that by adding BMP7 to TGFβ1-supplemented induction medium, ESC-MSC chondrogenesis is upregulated through increased production of endogenous TGFβ and activities of TGFβ and BMP signaling. J. Cell. Biochem. 118: 172-181, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Patrick T Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin.,Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
12
|
Pengjam Y, Madhyastha H, Madhyastha R, Yamaguchi Y, Nakajima Y, Maruyama M. Anthraquinone Glycoside Aloin Induces Osteogenic Initiation of MC3T3-E1 Cells: Involvement of MAPK Mediated Wnt and Bmp Signaling. Biomol Ther (Seoul) 2016; 24:123-31. [PMID: 26869456 PMCID: PMC4774492 DOI: 10.4062/biomolther.2015.106] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/10/2015] [Accepted: 10/21/2015] [Indexed: 01/31/2023] Open
Abstract
Osteoporosis is a bone pathology leading to increased fracture risk and challenging the quality of life. The aim of this study was to evaluate the effect of an anthraquinone glycoside, aloin, on osteogenic induction of MC3T3-E1 cells. Aloin increased alkaline phosphatase (ALP) activity, an early differentiation marker of osteoblasts. Aloin also increased the ALP activity in adult human adipose-derived stem cells (hADSC), indicating that the action of aloin was not cell-type specific. Alizarin red S staining revealed a significant amount of calcium deposition in cells treated with aloin. Aloin enhanced the expression of osteoblast differentiation genes, Bmp-2, Runx2 and collagen 1a, in a dose-dependent manner. Western blot analysis revealed that noggin and inhibitors of p38 MAPK and SAPK/JNK signals attenuated aloin-promoted expressions of Bmp-2 and Runx2 proteins. siRNA mediated blocking of Wnt-5a signaling pathway also annulled the influence of aloin, indicating Wnt-5a dependent activity. Inhibition of the different signal pathways abrogated the influence of aloin on ALP activity, confirming that aloin induced MC3T3-E1 cells into osteoblasts through MAPK mediated Wnt and Bmp signaling pathway.
Collapse
Affiliation(s)
- Yutthana Pengjam
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan.,Faculty of Medical Technology, Prince of Songkla University, HatYai, Songkhla 90110, Thailand
| | - Harishkumar Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Radha Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuya Yamaguchi
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuichi Nakajima
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Masugi Maruyama
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
13
|
Hettiaratchi MH, Guldberg RE, McDevitt TC. Biomaterial strategies for controlling stem cell fate via morphogen sequestration. J Mater Chem B 2016; 4:3464-3481. [DOI: 10.1039/c5tb02575c] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review explores the role of protein sequestration in the stem cell niche and how it has inspired the design of biomaterials that exploit natural protein sequestration to influence stem cell fate.
Collapse
Affiliation(s)
- M. H. Hettiaratchi
- The Parker H. Petit Institute for Bioengineering and Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
- The Wallace H. Coulter Department of Biomedical Engineering
| | - R. E. Guldberg
- The Parker H. Petit Institute for Bioengineering and Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
- The George W. Woodruff School of Mechanical Engineering
| | - T. C. McDevitt
- The Gladstone Institute of Cardiovascular Disease
- San Francisco
- USA
- The Department of Bioengineering and Therapeutic Sciences
- University of California San Francisco
| |
Collapse
|
14
|
Bulycheva E, Rauner M, Medyouf H, Theurl I, Bornhäuser M, Hofbauer LC, Platzbecker U. Myelodysplasia is in the niche: novel concepts and emerging therapies. Leukemia 2014; 29:259-68. [PMID: 25394715 PMCID: PMC4320287 DOI: 10.1038/leu.2014.325] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/25/2014] [Indexed: 12/28/2022]
Abstract
Myelodysplastic syndromes (MDSs) represent clonal disorders mainly of the elderly that are characterized by ineffective hematopoiesis and an increased risk of transformation into acute myeloid leukemia. The pathogenesis of MDS is thought to evolve from accumulation and selection of specific genetic or epigenetic events. Emerging evidence indicates that MDS is not solely a hematopoietic disease but rather affects the entire bone marrow microenvironment, including bone metabolism. Many of these cells, in particular mesenchymal stem and progenitor cells (MSPCs) and osteoblasts, express a number of adhesion molecules and secreted factors that regulate blood regeneration throughout life by contributing to hematopoietic stem and progenitor cell (HSPC) maintenance, self-renewal and differentiation. Several endocrine factors, such as erythropoietin, parathyroid hormone and estrogens, as well as deranged iron metabolism modulate these processes. Thus, interactions between MSPC and HSPC contribute to the pathogenesis of MDS and associated pathologies. A detailed understanding of these mechanisms may help to define novel targets for diagnosis and possibly therapy. In this review, we will discuss the scientific rationale of ‘osteohematology' as an emerging research field in MDS and outline clinical implications.
Collapse
Affiliation(s)
- E Bulycheva
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - M Rauner
- Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - H Medyouf
- Georg-Speyer-Haus, Institut for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - I Theurl
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - M Bornhäuser
- 1] Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - L C Hofbauer
- 1] Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - U Platzbecker
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
15
|
Tang Z, Wang Z, Qing F, Ni Y, Fan Y, Tan Y, Zhang X. Bone morphogenetic protein Smads signaling in mesenchymal stem cells affected by osteoinductive calcium phosphate ceramics. J Biomed Mater Res A 2014; 103:1001-10. [DOI: 10.1002/jbm.a.35242] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 05/19/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Zhurong Tang
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Zhe Wang
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Fangzhu Qing
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Yilu Ni
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Yanfei Tan
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials; Sichuan University; Chengdu 610064 China
| |
Collapse
|
16
|
Biver E, Thouverey C, Magne D, Caverzasio J. Crosstalk between tyrosine kinase receptors, GSK3 and BMP2 signaling during osteoblastic differentiation of human mesenchymal stem cells. Mol Cell Endocrinol 2014; 382:120-130. [PMID: 24060635 DOI: 10.1016/j.mce.2013.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/16/2013] [Accepted: 09/16/2013] [Indexed: 10/26/2022]
Abstract
Bone morphogenic proteins (BMPs) promote mesenchymal stem cell (MSC) osteogenic differentiation, whereas platelet derived growth factor (PDGF) and fibroblast growth factor (FGF) activate their proliferation through receptors tyrosine kinase (RTK). The effects of PDGF or FGF receptor signaling pathway on BMP2-induced osteoblastic differentiation was investigated in human MSC (HMSC). Inhibition of PDGF or/and FGF receptors enhanced BMP2-induced alkaline phosphatase (ALP) activity, expression of Osterix, ALP and Bone sialoprotein, and matrix calcification. These effects were associated with increased Smad-1 activity, indicating that mitogenic factors interfere with Smad signaling in HMSC differentiation. RTK activate MAPK and inhibit GSK3 through the PI3K/Akt pathway. Biochemical analysis indicated that MAPK JNK and GSK3 especially are potential signaling molecules regulating BMP-induced osteoblastic HMSC differentiation. These observations highlight that the osteogenic effects of BMP2 are modulated by mitogenic factors acting through RTK.
Collapse
Affiliation(s)
- Emmanuel Biver
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland; Pathophysiology of Inflammatory Bone Diseases, PMOI EA4490, Boulogne/Mer, France
| | - Cyril Thouverey
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland
| | - David Magne
- Institut of Molecular and Supramolecular Biochemistry, UMR, CNRS 5246, University of Lyon 1, 69622 Villeurbanne Cedex, France
| | - Joseph Caverzasio
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland.
| |
Collapse
|
17
|
Lui PPY. Histopathological changes in tendinopathy--potential roles of BMPs? Rheumatology (Oxford) 2013; 52:2116-2126. [DOI: 10.1093/rheumatology/ket165] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
18
|
Dai J, Li Y, Zhou H, Chen J, Chen M, Xiao Z. Genistein promotion of osteogenic differentiation through BMP2/SMAD5/RUNX2 signaling. Int J Biol Sci 2013; 9:1089-98. [PMID: 24339730 PMCID: PMC3858582 DOI: 10.7150/ijbs.7367] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 10/30/2013] [Indexed: 02/06/2023] Open
Abstract
To investigate the effects of Genistein on the osteogenic related gene expression profiles during osteoblastic differentiation of human bone marrow mesenchymal stem cell (hBMSC) cultures, the hBMSCs were cultured under osteogenic differentiation medium with the addition of Genistein (10(-8)∼10(-5) M) for 12 days. The cell proliferation was measured by BrdU incorporation, while the osteoblastic differentiation in hBMSC cultures was assessed by cellular alkaline phosphatase (ALP) activity. The cell apoptosis was determined by caspase 3/7 activation. GEArray Q series human osteogenesis gene array was used to analyze large-scale gene expression in Genistein-treated hBMSC cultures compared to the control group. Quantitative real-time RT-PCR, small interfering RNA (siRNA), and western blot analysis were used to confirm the microarray data in five representative transcripts. Genistein (10(-8)∼10(-6) M) dose- and time-dependently increased cell proliferation and cellular ALP activity, but had no significant effect on cell apoptosis in hBMSC cultures. The 96-gene array analysis indicated that 22 genes were upregulated more than 2-fold and 7 genes were downregulated at least 1.5-fold. The expressions of bone morphogenetic proteins (BMPs), small mothers against decapentaplegic homologs (SMADs), and Runt-related transcription factor 2 (RUNX2) were concomitantly increased under Genistein treatment while insulin-like growth factor 2 and inhibitory SMADs 6 and 7 expressions were significantly decreased. The results of the real-time RT-PCR had a correlation with the results of microarray analysis and were estrogen-receptor dependent. Specific gene siRNAs knock-down further confirmed the osteogenic effects of Genistein on BMP2, SMAD5 and RUNX2 protein expression. Genistein enhanced osteogenic differentiation in cultured hBMSCs mainly through the BMP-dependent SMADs and RUNX2 signaling.
Collapse
Affiliation(s)
- Jin Dai
- 1. Division of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China; ; 2. Institute of Clinical Pharmacology, Xiangya Medical College, Central South University, Changsha, Hunan, 410078, China
| | | | | | | | | | | |
Collapse
|
19
|
Lai W, Li Y, Mak S, Ho F, Chow S, Chooi W, Chow C, Leung A, Chan B. Reconstitution of bone-like matrix in osteogenically differentiated mesenchymal stem cell-collagen constructs: A three-dimensional in vitro model to study hematopoietic stem cell niche. J Tissue Eng 2013; 4:2041731413508668. [PMID: 24555007 PMCID: PMC3927755 DOI: 10.1177/2041731413508668] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/19/2013] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and osteoblasts are important niche cells for hematopoietic stem cells (HSCs) in bone marrow osteoblastic niche. Here, we aim to partially reconstitute the bone marrow HSC niche in vitro using collagen microencapsulation for investigation of the interactions between HSCs and MSCs. Mouse MSCs (mMSCs) microencapsulated in collagen were osteogenically differentiated to derive a bone-like matrix consisting of osteocalcin, osteopontin, and calcium deposits and secreted bone morphogenic protein 2 (BMP2). Decellularized bone-like matrix was seeded with fluorescence-labeled human MSCs and HSCs. Comparing with pure collagen scaffold, significantly more HSCs and HSC–MSC pairs per unit area were found in the decellularized bone-like matrix. Moreover, incubation with excess neutralizing antibody of BMP2 resulted in a significantly higher number of HSC per unit area than that without in the decellularized matrix. This work suggests that the osteogenic differentiated MSC–collagen microsphere is a valuable three-dimensional in vitro model to elucidate cell–cell and cell–matrix interactions in HSC niche.
Collapse
Affiliation(s)
- Wy Lai
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Yy Li
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Sk Mak
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Fc Ho
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - St Chow
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Wh Chooi
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Ch Chow
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ay Leung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Bp Chan
- Tissue Engineering Laboratory, Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| |
Collapse
|
20
|
Rivera JC, Strohbach CA, Wenke JC, Rathbone CR. Beyond osteogenesis: an in vitro comparison of the potentials of six bone morphogenetic proteins. Front Pharmacol 2013; 4:125. [PMID: 24101902 PMCID: PMC3787247 DOI: 10.3389/fphar.2013.00125] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/09/2013] [Indexed: 12/21/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) other than the clinically available BMP-2 and BMP-7 may be useful for improving fracture healing through both increasing osteogenesis and creating a favorable healing environment by altering cytokine release by endogenous cells. Given the spectrum of potential applications for BMPs, the objective of this study was to evaluate various BMPs under a variety of conditions to provide further insight into their therapeutic capabilities. The alkaline phosphatase (ALP) activity of both C2C12 and human adipose-derived stem cells (hASCs) was measured after exposure of increasing doses of recombinant human BMP-2, -4, -5, -6, -7, or -9 for 3 and 7 days. BMPs-2, -4, -5, -6, -7, and -9 were compared in terms of their ability to affect the release of stromal derived factor-1 (SDF-1), vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (b-FGF) from human bone marrow stromal cells (hBMSCs). Gene expression of ALP, osteocalcin, SDF-1, VEGF, and b-FGF following shRNA-mediated knockdown of BMP-2 and BMP-6 in hBMSCs or human osteoblasts under osteogenic differentiation conditions was also evaluated. Collectively, BMPs-6 and -9 produced the greatest osteogenic differentiation of C2C12 and hASCs as determined by ALP. The hBMSC secretion of SDF-1 was most affected by BMP-5, VEGF by BMP-4, and b-FGF by BMP-2. The knockdown of BMP-2 in BMSCs had no effect on any of the genes measured whereas BMP-6 knockdown in hBMSCs caused a significant increase in VEGF gene expression. BMP-2 and BMP-6 knockdown in human osteoblasts caused significant increases in VEGF gene expression and trends toward decreases in osteocalcin expression. These findings support efforts to study other BMPs as potential bone graft supplements, and to consider combined BMP delivery for promotion of multiple aspects of fracture healing.
Collapse
Affiliation(s)
- Jessica C Rivera
- Extremity Trauma and Regenerative Medicine, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | | | | | | |
Collapse
|
21
|
Cruz ACC, Silva ML, Caon T, Simões CMO. Addition of bone morphogenetic protein type 2 to ascorbate and β-glycerophosphate supplementation did not enhance osteogenic differentiation of human adipose-derived stem cells. J Appl Oral Sci 2013; 20:628-35. [PMID: 23329244 PMCID: PMC3881851 DOI: 10.1590/s1678-77572012000600007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 09/14/2012] [Indexed: 12/25/2022] Open
Abstract
Bone morphogenetic protein type 2 (BMP-2) is a potent local factor, which promotes
bone formation and has been used as an osteogenic supplement for mesenchymal stem
cells.
Collapse
|
22
|
Huang YZ, Cai JQ, Lv FJ, Xie HL, Yang ZM, Huang YC, Deng L. Species variation in the spontaneous calcification of bone marrow-derived mesenchymal stem cells. Cytotherapy 2013; 15:323-329. [DOI: 10.1016/j.jcyt.2012.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 10/10/2012] [Accepted: 11/13/2012] [Indexed: 01/13/2023]
|
23
|
Aizman I, McGrogan M, Case CC. Quantitative microplate assay for studying mesenchymal stromal cell-induced neuropoiesis. Stem Cells Transl Med 2013; 2:223-32. [PMID: 23430693 DOI: 10.5966/sctm.2012-0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplanting mesenchymal stromal cells (MSCs) or their derivatives in a neurodegenerative environment is believed to be beneficial because of the trophic support, migratory guidance, and neurogenic stimuli they provide. There is a growing need for in vitro models of mesenchymal-neural cell interactions to enable identification of mediators of the MSC activity and quantitative assessment of neuropoietic potency of MSC preparations. Here, we characterize a microplate-format coculture system in which primary embryonic rat cortex cells are directly cocultured with human MSCs on cell-derived extracellular matrix (ECM) in the absence of exogenous growth factors. In this system, expression levels of the rat neural stem/early progenitor marker nestin, as well as neuronal and astrocytic markers, directly depended on MSC dose, whereas an oligodendrogenic marker exhibited a biphasic MSC-dose response, as measured using species-specific quantitative reverse transcription-polymerase chain reaction in total cell lysates and confirmed using immunostaining. Both neural cell proliferation and differentiation contributed to the MSC-mediated neuropoiesis. ECM's heparan sulfate proteoglycans were essential for the growth of the nestin-positive cell population. Neutralization studies showed that MSC-derived fibroblast growth factor 2 was a major and diffusible inducer of rat nestin, whereas MSC-derived bone morphogenetic proteins (BMPs), particularly, BMP4, were astrogenesis mediators, predominantly acting in a coculture setting. This system enables analysis of multifactorial MSC-neural cell interactions and can be used for elucidating the neuropoietic potency of MSCs and their derivative preparations.
Collapse
|
24
|
Büttner M, Möller S, Keller M, Huster D, Schiller J, Schnabelrauch M, Dieter P, Hempel U. Over-sulfated chondroitin sulfate derivatives induce osteogenic differentiation of hMSC independent of BMP-2 and TGF-β1 signalling. J Cell Physiol 2013; 228:330-40. [PMID: 22718137 DOI: 10.1002/jcp.24135] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Natural glycosaminoglycans (GAGs) and chemically modified GAG derivatives are known to support osteogenic differentiation of mesenchymal stromal cells (MSC). This effect has mainly been described to be mediated by increasing the effectiveness of bone anabolic growth factors such as bone morphogenetic proteins (BMPs) due to the binding and presentation of the growth factor or by modulating its signal transduction pathway. In the present study, the influence of chondroitin sulfate (CS) and two chemically over-sulfated CS derivatives on osteogenic differentiation of human mesenchymal stromal cells (hMSC) and on BMP-2 and transforming growth factor β1 (TGF-β1) signalling was investigated. Over-sulfated CS derivatives induced an increase of tissue non-specific alkaline phosphatase (TNAP) activity and calcium deposition, whereas collagen synthesis was slightly decreased. The BMP-2-induced Smad1/5 activation was inhibited in the presence of over-sulfated CS derivatives leading to a loss of BMP-2-induced TNAP activity and calcium deposition. In contrast, the TGF-β1-induced activation of Smad2/3 and collagen synthesis were not affected by the over-sulfated CS derivatives. BMP-2 and TGF-β1 did not activate the extracellular signal-regulated kinase 1/2 or mitogen-activated protein kinase p38 in hMSC. These data suggest that over-sulfated CS derivatives themselves are able to induce osteogenic differentiation, probably independent of BMP-2 and TGF-β1 signalling, and offer therefore an interesting approach for the improvement of bone healing.
Collapse
Affiliation(s)
- Marianne Büttner
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Biver E, Soubrier AS, Thouverey C, Cortet B, Broux O, Caverzasio J, Hardouin P. Fibroblast growth factor 2 inhibits up-regulation of bone morphogenic proteins and their receptors during osteoblastic differentiation of human mesenchymal stem cells. Biochem Biophys Res Commun 2012; 427:737-42. [PMID: 23044416 DOI: 10.1016/j.bbrc.2012.09.129] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Accepted: 09/27/2012] [Indexed: 12/30/2022]
Abstract
Understanding the interactions between growth factors and bone morphogenic proteins (BMPs) signaling remains a crucial issue to optimize the use of human mesenchymal stem cells (HMSCs) and BMPs in therapeutic perspectives and bone tissue engineering. BMPs are potent inducers of osteoblastic differentiation. They exert their actions via BMP receptors (BMPR), including BMPR1A, BMPR1B and BMPR2. Fibroblast growth factor 2 (FGF2) is expressed by cells of the osteoblastic lineage, increases their proliferation and is secreted during the healing process of fractures or in surgery bone sites. We hypothesized that FGF2 might influence HMSC osteoblastic differentiation by modulating expressions of BMPs and their receptors. BMP2, BMP4, BMPR1A and mainly BMPR1B expressions were up-regulated during this differentiation. FGF2 inhibited HMSCs osteoblastic differentiation and the up-regulation of BMPs and BMPR. This effect was prevented by inhibiting the ERK or JNK mitogen-activated protein kinases which are known to be activated by FGF2. These data provide a mechanism explaining the inhibitory effect of FGF2 on osteoblastic differentiation of HMSCs. These crosstalks between growth and osteogenic factors should be considered in the use of recombinant BMPs in therapeutic purpose of fracture repair or skeletal bioengineering.
Collapse
Affiliation(s)
- Emmanuel Biver
- Physiopathology of Inflammatory Bone Diseases, EA 4490, University Lille North of France, Quai Masset, Bassin Napoléon, BP120, 62327 Boulogne sur Mer, France.
| | | | | | | | | | | | | |
Collapse
|
26
|
Lescarbeau RM, Seib FP, Prewitz M, Werner C, Kaplan DL. In vitro model of metastasis to bone marrow mediates prostate cancer castration resistant growth through paracrine and extracellular matrix factors. PLoS One 2012; 7:e40372. [PMID: 22870197 PMCID: PMC3411611 DOI: 10.1371/journal.pone.0040372] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/07/2012] [Indexed: 11/26/2022] Open
Abstract
The spread of prostate cancer cells to the bone marrow microenvironment and castration resistant growth are key steps in disease progression and significant sources of morbidity. However, the biological significance of mesenchymal stem cells (MSCs) and bone marrow derived extracellular matrix (BM-ECM) in this process is not fully understood. We therefore established an in vitro engineered bone marrow tissue model that incorporates hMSCs and BM-ECM to facilitate mechanistic studies of prostate cancer cell survival in androgen-depleted media in response to paracrine factors and BM-ECM. hMSC-derived paracrine factors increased LNCaP cell survival, which was in part attributed to IGFR and IL6 signaling. In addition, BM-ECM increased LNCaP and MDA-PCa-2b cell survival in androgen-depleted conditions, and induced chemoresistance and morphological changes in LNCaPs. To determine the effect of BM-ECM on cell signaling, the phosphorylation status of 46 kinases was examined. Increases in the phosphorylation of MAPK pathway-related proteins as well as sustained Akt phosphorylation were observed in BM-ECM cultures when compared to cultures grown on plasma-treated polystyrene. Blocking MEK1/2 or the PI3K pathway led to a significant reduction in LNCaP survival when cultured on BM-ECM in androgen-depleted conditions. The clinical relevance of these observations was determined by analyzing Erk phosphorylation in human bone metastatic prostate cancer versus non-metastatic prostate cancer, and increased phosphorylation was seen in the metastatic samples. Here we describe an engineered bone marrow model that mimics many features observed in patients and provides a platform for mechanistic in vitro studies.
Collapse
Affiliation(s)
- Reynald M. Lescarbeau
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - F. Philipp Seib
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Marina Prewitz
- Leibniz Institute for Polymer Research Dresden, Dresden, Germany
| | - Carsten Werner
- Leibniz Institute for Polymer Research Dresden, Dresden, Germany
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
27
|
Kim J, Ma T. Bioreactor strategy in bone tissue engineering: pre-culture and osteogenic differentiation under two flow configurations. Tissue Eng Part A 2012; 18:2354-64. [PMID: 22690750 DOI: 10.1089/ten.tea.2011.0674] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Since robust osteogenic differentiation and mineralization are integral to the engineering of bone constructs, understanding the impact of the cellular microenvironments on human mesenchymal stem cell (hMSCs) osteogenic differentiation is crucial to optimize bioreactor strategy. Two perfusion flow conditions were utilized in order to understand the impact of the flow configuration on hMSC construct development during both pre-culture (PC) in growth media and its subsequent osteogenic induction (OI). The media in the in-house perfusion bioreactor was controlled to perfuse either around (termed parallel flow [PF]) the construct surfaces or penetrate through the construct (termed transverse flow [TF]) for 7 days of the PC followed by 7 days of the OI. The flow configuration during the PC not only changed growth kinetics but also influenced cell distribution and potency of osteogenic differentiation and mineralization during the subsequent OI. While shear stress resulted from the TF stimulated cell proliferation during PC, the convective removal of de novo extracellular matrix (ECM) proteins and growth factors (GFs) reduced cell proliferation on OI. In contrast, the effective retention of de novo ECM proteins and GFs in the PC constructs under the PF maintained cell proliferation under the OI but resulted in localized cell aggregations, which influenced their osteogenic differentiation. The results revealed the contrasting roles of the convective flow as a mechanical stimulus, the redistribution of the cells and macromolecules in 3D constructs, and their divergent impacts on cellular events, leading to bone construct formation. The results suggest that the modulation of the flow configuration in the perfusion bioreactor is an effective strategy that regulates the construct properties and maximizes the functional outcome.
Collapse
Affiliation(s)
- Junho Kim
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida 32310, USA
| | | |
Collapse
|
28
|
Rui YF, Lui PPY, Rolf CG, Wong YM, Lee YW, Chan KM. Expression of chondro-osteogenic BMPs in clinical samples of patellar tendinopathy. Knee Surg Sports Traumatol Arthrosc 2012; 20:1409-1417. [PMID: 21946950 DOI: 10.1007/s00167-011-1685-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 09/13/2011] [Indexed: 01/07/2023]
Abstract
PURPOSE The pathogenesis of patellar tendinopathy remains unclear. Expression of BMP-2/-4/-7 was reported in an ossified failed tendon healing animal model of patellar tendinopathy. This study aimed to investigate the expression of these chondro-osteogenic BMPs in clinical samples of patellar tendinopathy. METHODS Patellar tendon samples were collected from 16 consecutive patients with patellar tendinopathy and 16 consecutive controls undergoing anterior cruciate ligament reconstruction with bone-patellar tendon-bone autograft in the authors' hospital after getting their consent. The expression of BMP-2/-4/-7 was examined in all samples using immunohistochemistry. Ossification observed in two tendinopathy samples was characterized by histology, alizarin red S staining, alcian blue staining, TRAP staining and immunohistochemical staining of Sox9, osteopontin (OPN) and osteocalcin (OCN). RESULTS Regions of hypo- and hyper-cellularity and vascularity, with loss of crimp structure of collagen matrix, were observed in patellar tendinopathy samples. Round cells and in some cases, cells with typical chondrocyte phenotype were observed. For the ossified tendinopathy samples with positive alizarin red S staining, OPN-positive and Sox9-positive chondrocyte-like cells in alcian blue-stained extracellular matrix, OCN-positive osteoblast-like cells and TRAP-positive multi-nucleated cells were observed around the ossified deposits. No expression of BMP-2/-4/-7 was observed in healthy patellar tendons. However, the expression of BMP-2/-4/-7 was observed in all patellar tendinopathy samples with or without ossification. CONCLUSIONS Clinical samples of patellar tendinopathy showed ectopic expression of BMP-2/-4/-7. This was not evident in control samples from healthy patellar tendons. LEVEL OF EVIDENCE Prognostic studies, Level III.
Collapse
Affiliation(s)
- Yun Feng Rui
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
29
|
Chiang ZC, Yu SH, Chao AC, Dong GC. Preparation and characterization of dexamethasone-immobilized chitosan scaffold. J Biosci Bioeng 2012; 113:654-60. [DOI: 10.1016/j.jbiosc.2012.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 12/27/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
|
30
|
Rui YF, Lui PPY, Lee YW, Chan KM. Higher BMP receptor expression and BMP-2-induced osteogenic differentiation in tendon-derived stem cells compared with bone-marrow-derived mesenchymal stem cells. INTERNATIONAL ORTHOPAEDICS 2012; 36:1099-1107. [PMID: 22134708 PMCID: PMC3337107 DOI: 10.1007/s00264-011-1417-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 11/06/2011] [Indexed: 01/30/2023]
Abstract
PURPOSE Surgical reattachment of tendon to bone often fails due to regeneration failure of the specialised tendon-bone junction (TBJ). The use of mesenchymal stem cells for TBJ regeneration has been reported with promising results. Tendon-derived stem cells (TDSCs) with high proliferative and multi-lineage differentiation potential have been isolated. As stem cells residing in tendons, TDSCs can be considered a new cell source for TBJ repair. Bone morphogenic protein 2 (BMP-2) is a potent osteogenic factor with roles in normal bone healing and pathological ectopic bone formation in soft tissues. The use of BMP-2 to promote TBJ repair has been well reported. This study aimed to compare TDSCs to the gold standard bone-marrow-derived mesenchymal stem cells (BMSCs) with respect to osteogenic response to BMP-2 in vitro. METHOD The clonogenicity and multi-differentiation potential of TDSCs and BMSCs were identified by colony-forming-unit assay, osteogenic, adipogenic and chondrogenic differentiation assays. Their osteogenic response to BMP-2 in vitro was examined by alkaline phosphatase (ALP) cytochemical staining, ALP activity assay and Alizarin red S staining of calcium nodule formation. Messenger RNA (mRNA) and BMP receptor (types IA, IB and II) protein expression were examined by quantitative real-time reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blotting. RESULTS Our results showed that both TDSCs and BMSCs exhibited stem cell properties, including clonogenicity and multi-differentiation potential. TDSCs expressed higher mRNA and protein levels of BMP receptors IA, IB and II. They also exhibited higher osteogenic differentiation with and without BMP-2 stimulation compared with BMSCs. CONCLUSIONS TDSCs with/without BMP-2 might be an attractive source for TBJ repair compared with BMSCs.
Collapse
Affiliation(s)
- Yun Feng Rui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Rm. 74025, 5/F, Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong SAR China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
- Department of Orthopaedics, Zhongda Hospital, Southeast University, 87 Ding Jia Qiao, Nanjing, Jiangsu 210009 China
| | - Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Rm. 74025, 5/F, Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong SAR China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
- Program of Stem Cell and Regeneration, School of Biomedical Science, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| | - Yuk Wai Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Rm. 74025, 5/F, Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong SAR China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| | - Kai Ming Chan
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Rm. 74025, 5/F, Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong SAR China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
- Program of Stem Cell and Regeneration, School of Biomedical Science, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| |
Collapse
|
31
|
Lui PPY, Chan KM. Tendon-derived stem cells (TDSCs): from basic science to potential roles in tendon pathology and tissue engineering applications. Stem Cell Rev Rep 2012; 7:883-97. [PMID: 21611803 DOI: 10.1007/s12015-011-9276-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traditionally, tendons are considered to only contain tenocytes that are responsible for the maintenance, repair and remodeling of tendons. Stem cells, which are termed tendon-derived stem cells (TDSCs), have recently been identified in tendons. This review aims to summarize the current information about the in vitro characteristics of TDSCs, including issues related to TDSC isolation and culture, their cell morphology, immunophenotypes, proliferation and differentiation characteristics and senescence during in vitro passaging. The challenges in studying the functions of these cells are also discussed. The niche where TDSCs resided essentially provides signals that are conducive to the maintenance of definitive stem cell properties of TDSCs. Yet the niche may also induce pathologies by imposing an aberrant function on TDSCs or other targets. The possible niche factors of TDSCs are herein discussed. We presented current evidences supporting the potential pathogenic role of TDSCs in the development of tendinopathy with reference to the recent findings on the altered biological responses of these cells in response to their potential niche factors. The use of resident stem cells may promote engraftment and differentiation of transplanted cells in tendon and tendon-bone junction repair because the tendon milieu is an ideal and familiar environment to the transplanted cells. Evidences are presented to show the potential advantages and results of using TDSCs as a new cell source for tendon and tendon-bone junction repair. Issues pertaining to the use of TDSCs for tissue repair are also discussed.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| | | |
Collapse
|
32
|
Siegel G, Krause P, Wöhrle S, Nowak P, Ayturan M, Kluba T, Brehm BR, Neumeister B, Köhler D, Rosenberger P, Just L, Northoff H, Schäfer R. Bone marrow-derived human mesenchymal stem cells express cardiomyogenic proteins but do not exhibit functional cardiomyogenic differentiation potential. Stem Cells Dev 2012; 21:2457-70. [PMID: 22309203 DOI: 10.1089/scd.2011.0626] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Despite their paracrine activites, cardiomyogenic differentiation of bone marrow (BM)-derived mesenchymal stem cells (MSCs) is thought to contribute to cardiac regeneration. To systematically evaluate the role of differentiation in MSC-mediated cardiac regeneration, the cardiomyogenic differentiation potential of human MSCs (hMSCs) and murine MSCs (mMSCs) was investigated in vitro and in vivo by inducing cardiomyogenic and noncardiomyogenic differentiation. Untreated hMSCs showed upregulation of cardiac tropopin I, cardiac actin, and myosin light chain mRNA and protein, and treatment of hMSCs with various cardiomyogenic differentiation media led to an enhanced expression of cardiomyogenic genes and proteins; however, no functional cardiomyogenic differentiation of hMSCs was observed. Moreover, co-culturing of hMSCs with cardiomyocytes derived from murine pluripotent cells (mcP19) or with murine fetal cardiomyocytes (mfCMCs) did not result in functional cardiomyogenic differentiation of hMSCs. Despite direct contact to beating mfCMCs, hMSCs could be effectively differentiated into cells of only the adipogenic and osteogenic lineage. After intramyocardial transplantation into a mouse model of myocardial infarction, Sca-1(+) mMSCs migrated to the infarcted area and survived at least 14 days but showed inconsistent evidence of functional cardiomyogenic differentiation. Neither in vitro treatment nor intramyocardial transplantation of MSCs reliably generated MSC-derived cardiomyocytes, indicating that functional cardiomyogenic differentiation of BM-derived MSCs is a rare event and, therefore, may not be the main contributor to cardiac regeneration.
Collapse
Affiliation(s)
- Georg Siegel
- Institute of Clinical and Experimental Transfusion Medicine (IKET), University Hospital Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rashidi H, Strohbuecker S, Jackson L, Kalra S, Blake AJ, France L, Tufarelli C, Sottile V. Differences in the pattern and regulation of mineral deposition in human cell lines of osteogenic and non-osteogenic origin. Cells Tissues Organs 2011; 195:484-94. [PMID: 22123583 DOI: 10.1159/000329861] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2011] [Indexed: 12/21/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are widely used as a cellular model of bone formation, and can mineralize in vitro in response to osteogenic medium (OM). It is unclear, however, whether this property is specific to cells of mesenchymal origin. We analysed the OM response in 3 non-osteogenic lines, HEK293, HeLa and NTera, compared to MSCs. Whereas HEK293 cells failed to respond to OM conditions, the 2 carcinoma-derived lines NTera and HeLa deposited a calcium phosphate mineral comparable to that present in MSC cultures. However, unlike MSCs, HeLa and NTera cultures did so in the absence of dexamethasone. This discrepancy was confirmed, as bone morphogenetic protein inhibition obliterated the OM response in MSCs but not in HeLa or NTera, indicating that these 2 models can deposit mineral through a mechanism independent of established dexamethasone or bone morphogenetic protein signalling.
Collapse
Affiliation(s)
- Hassan Rashidi
- School of Clinical Sciences, Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, The University of Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
34
|
He J, Dong J, Wang T, Xu H, Dai C, Ma S, Zhu L. Bone morphogenetic protein receptor IB as a marker for enrichment of osteogenic precursor-like cells in human dermis. Arch Dermatol Res 2011; 303:581-90. [PMID: 21644047 DOI: 10.1007/s00403-011-1156-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/13/2011] [Accepted: 05/24/2011] [Indexed: 10/18/2022]
Abstract
The scarcity of bone marrow mesenchymal stromal cells (BMSCs) prompts the search for alternative sources for cell-based bone defects repair. Human dermal fibroblasts (FBs) have been shown to have a high proliferative potential and the capacity to differentiate into an osteogenic phenotype. The easy and repeated harvest in large quantities makes this cell source a potential candidate for bone tissue engineering. The aim of our study was to compare directly the immune phenotype, proliferative capacity and osteogenic differentiation potential of FBs with that of "gold standard" BMSCs or adipose-derived mesenchymal stromal cells (ADSCs), another alternative osteoprogenitor cell source. Flow cytometry demonstrated that FBs, ADSCs and BMSCs shared common cell surface marker protein expression profiles when using a panel of surface antigens. FBs had the highest proliferative potential, but lowest osteogenic differentiation potential in vitro, compared with ADSCs or BMSCs. More importantly, BMPR-IB(+)-sorted FBs subpopulation had a higher osteogenic differentiation potential than BMPR-IB(-)-sorted FBs subpopulation. Our results indicated that the heterogeneous FBs were not an appropriate cell source for bone tissue engineering. Immunoselection by BMPR-IB can generate highly purified osteogenic precursor-like cells in the human dermis.
Collapse
Affiliation(s)
- Jinguang He
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Jiao Tong University School of Medicine, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
35
|
Yee Lui PP, Wong YM, Rui YF, Lee YW, Chan LS, Chan KM. Expression of chondro-osteogenic BMPs in ossified failed tendon healing model of tendinopathy. J Orthop Res 2011; 29:816-821. [PMID: 21520255 DOI: 10.1002/jor.21313] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 10/08/2010] [Indexed: 02/04/2023]
Abstract
Chondrocytes phenotype/markers were expressed in clinical samples of tendinopathy and calcifying tendinopathy. This study examined the spatial-temporal expression of chondro-osteogenic Bone Morphogenetic Proteins (BMPs), which might contribute to ectopic chondro-osteogenesis and failed healing process in tendinopathy. Collagenase was injected into patellar tendon of rats to induce ossified failed tendon healing. At week 2, 4, 8, 12, and 16, the patella tendon was harvested for immunohistochemical staining and analysis of BMP-2/4/7. BMP-4/7 showed similar expression patterns, which was different from BMP-2. The expression of BMP-2 in the tendon matrix increased at week 2 and was reduced to nearly undetectable level afterwards except at the chondro-ossification sites. However, the expression of BMP-4/7 in the healing tendon fibroblast-like cells and matrix increased at week 2, reduced at week 4 and 8 and increased again at week 12 and 16, consistent with transient healing at week 8 in this animal model. There was increasing strong expression of BMP-4/7 in the chondrocyte-like cells in the un-ossified and ossified areas from week 8-16. BMP-4/7, besides BMP-2, might also contribute to ectopic chondro-osteogenesis and failed healing in tendon injuries. BMP-4/7, but not BMP-2, might be involved in regulating late events in ossified failed tendon healing.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | | | | | | | | | | |
Collapse
|
36
|
Seib FP, Lanfer B, Bornhäuser M, Werner C. Biological activity of extracellular matrix-associated BMP-2. J Tissue Eng Regen Med 2010; 4:324-7. [PMID: 20014079 DOI: 10.1002/term.240] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The critical requirement for matrix-associated bone morphogenetic proteins (BMPs) during induction of bone formation in vivo has long been recognized. However, the role of extracellular matrix (ECM) physisorbed BMPs in inducing the differentiation of resident mesenchymal stem cells into osteoblasts has been ill-defined. We therefore used BMP-responsive C2C12s to study the biological activity of collagen type I physisorbed BMP-2. Fibrillar collagen type I scaffolds were loaded with 75 ng BMP-2/microg collagen. Under cell culture conditions, 40% of loaded (125)I-labelled BMP-2 was released within 24 h, whereas the remaining BMP-2 was stably physisorbed for > 7 days. Using these systems suggested that physisorbed BMP-2 is more active than diffusible BMP-2. Thus, the current clinical practice of immobilizing BMPs on collagen type I scaffolds not only prolongs local delivery of the morphogen but could also enhance biological activity at the cellular level.
Collapse
Affiliation(s)
- F Philipp Seib
- Leibniz Institute for Polymer Research, Max Bergmann Centre for Biomaterials, Dresden, Germany
| | | | | | | |
Collapse
|
37
|
Solmesky L, Lefler S, Jacob-Hirsch J, Bulvik S, Rechavi G, Weil M. Serum free cultured bone marrow mesenchymal stem cells as a platform to characterize the effects of specific molecules. PLoS One 2010; 5. [PMID: 20844755 PMCID: PMC2937025 DOI: 10.1371/journal.pone.0012689] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 08/23/2010] [Indexed: 11/18/2022] Open
Abstract
Human mesenchymal stem cells (hMSC) are easily isolated from the bone marrow by adherence to plastic surfaces. These cells show self-renewal capacity and multipotency. A unique feature of hMSC is their capacity to survive without serum. Under this condition hMSC neither proliferate nor differentiate but maintain their biological properties unaffected. Therefore, this should be a perfect platform to study the biological effects of defined molecules on these human stem cells. We show that hMSC treated for five days with retinoic acid (RA) in the absence of serum undergo several transcriptional changes causing an inhibition of ERK related pathways. We found that RA induces the loss of hMSC properties such as differentiation potential to either osteoblasts or adipocytes. We also found that RA inhibits cell cycle progression in the presence of proliferating signals such as epidermal growth factor (EGF) combined with basic fibroblast growth factor (bFGF). In the same manner, RA showed to cause a reduction in cell adhesion and cell migration. In contrast to these results, the addition of EGF+bFGF to serum free cultures was enough to upregulate ERK activity and induce hMSC proliferation and cell migration. Furthermore, the addition of these factors to differentiation specific media instead of serum was enough to induce either osteogenesis or adipogenesis. Altogether, our results show that hMSC's ability to survive without serum enables the identification of signaling factors and pathways that are involved in their stem cell biological characteristics without possible serum interferences.
Collapse
Affiliation(s)
- Leonardo Solmesky
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Lefler
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Shlomo Bulvik
- Hematology Department, Laniado Hospital, Netanya, Israel
| | - Gideon Rechavi
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Miguel Weil
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
38
|
Payne KA, Meszaros LB, Phillippi JA, Huard J. Effect of phosphatidyl inositol 3-kinase, extracellular signal-regulated kinases 1/2, and p38 mitogen-activated protein kinase inhibition on osteogenic differentiation of muscle-derived stem cells. Tissue Eng Part A 2010; 16:3647-55. [PMID: 20617875 DOI: 10.1089/ten.tea.2009.0738] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Skeletal muscle-derived stem cells (MDSCs) can undergo osteogenesis when treated with bone morphogenetic proteins (BMPs), making them a potential cell source for bone tissue engineering. The signaling pathways that regulate BMP4-induced osteogenesis in MDSCs are not well understood, although they may provide a means to better regulate differentiation during bone regeneration. The objective of this study was to characterize the signaling pathways involved in the BMP4-induced osteogenesis of MDSCs. Cells were treated with BMP4 and specific inhibitors to the extracellular signal-regulated kinases 1/2 (ERK1/2), p38 mitogen-activated protein kinase (MAPK), and phosphatidyl inositol 3-kinase (PI3K) pathways (PD98059, SB203580, and Ly294002, respectively). Cellular proliferation, expression of osteoblast-related genes, alkaline phosphatase (ALP) activity, and tissue mineralization were measured to determine the role of each pathway in the osteogenic differentiation of MDSCs. Inhibition of the ERK1/2 pathway increased ALP activity and mineralization, whereas inhibition of the p38 MAPK pathway decreased osteogenesis, suggesting opposing roles of these pathways in the BMP4-induced osteogenesis of MDSCs. Inhibition of the PI3K pathway significantly increased mineralization by MDSCs. These findings highlight the involvement of the ERK1/2, p38 MAPK, and PI3K pathways in opposing capacities in MDSC differentiation and warrant further investigation, as it may identify novel therapeutic targets for the development of stem cell-based therapies for bone tissue engineering.
Collapse
Affiliation(s)
- Karin A Payne
- Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | | | | | | |
Collapse
|
39
|
Waris V, Waris E, Sillat T, Konttinen YT. BMPs in periprosthetic tissues around aseptically loosened total hip implants. Acta Orthop 2010; 81:420-6. [PMID: 20515435 PMCID: PMC2917563 DOI: 10.3109/17453674.2010.492765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Primary and dynamically maintained periprosthetic bone formation is essential for osseointegration of hip implants to host bone. Bone morphogenetic proteins (BMPs) play a role in osteoinductive bone formation. We hypothesized that there is an increased local synthesis of BMPs in the synovial membrane-like interface around aseptically loosened total hip replacement (THR) implants, as body attempts to generate or maintain implant fixation. PATIENTS AND METHODS We compared synovial membrane-like interface tissue from revised total hip replacements (rTHR, n = 9) to osteoarthritic control synovial membrane samples (OA, n = 11. Avidin-biotin-peroxidase complex staining and grading of BMP-2, BMP-4, BMP-6, and BMP-7 was done. Immunofluorescence staining was used to study BMP proteins produced by mesenchymal stromal/stem cells (MSCs) and osteoblasts. RESULTS AND INTERPRETATION All BMPs studied were present in the synovial lining or lining-like layer, fibroblast-like stromal cells, interstitial macrophage-like cells, and endothelial cells. In OA and rTHR samples, BMP-6 positivity in cells, inducible by the proinflammatory cytokines tumor necrosis factor-alpha and interleukin-1beta, predominated over expression of other BMPs. Macrophage-like cells positive for BMP-4, inducible in macrophages by stimulation with particles, were more frequent around loosened implants than in control OA samples, but apparently not enough to prevent loosening. MSCs contained BMP-2, BMP-4, BMP-6, and BMP-7, but this staining diminished during osteogenesis, suggesting that BMPs are produced by progenitor cells in particular, probably for storage in the bone matrix.
Collapse
Affiliation(s)
| | - Eero Waris
- Department of Hand Surgery, Helsinki University Central Hospital, Helsinki
| | | | | |
Collapse
|
40
|
Laflamme C, Curt S, Rouabhia M. Epidermal growth factor and bone morphogenetic proteins upregulate osteoblast proliferation and osteoblastic markers and inhibit bone nodule formation. Arch Oral Biol 2010; 55:689-701. [PMID: 20627196 DOI: 10.1016/j.archoralbio.2010.06.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 06/04/2010] [Accepted: 06/21/2010] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the in vitro osteogenic activity of EGF in association with bone morphogenetic proteins BMP2 and BMP7. METHODS SaOS-2 (osteoblast-like cell line from human osteosarcoma) were cultured in the presence of EGF and BMPs for various culture periods to assess (a) cell proliferation by MTT assay, (b) Runx2, alkaline phosphatase (ALP) and osteocalcin (OC) mRNA expression using quantitative RT-PCR and ELISA, and (c) bone tissue mineralization using Alizarin Red staining. RESULTS EGF alone was able to stimulate osteoblast growth in a time-dependent manner. When mixed with BMP2, BMP7, and their combination, EGF greatly promoted osteoblast growth, compared to the BMP- and EGF-stimulated cells, suggesting a possible synergistic effect between EGF and BMPs on osteoblast growth. Stimulation with EGF, EGF/BMP2, and EGF/BMP2/BMP7 for 7 days upregulated Runx2 mRNA expression by the osteoblasts. EGF downregulated ALP mRNA expression, which was recovered when the BMP2/BMP7 combination was added to the osteoblast culture. Tested on OC mRNA expression, EGF had no effect and inhibited the enhancing effect of BMP2 and BMP7 on osteocalcin expression. The bone mineralization assay showed that EGF reduced both the number and size of the bone nodules. This reducing effect was observable even in the presence of BMP2 and BMP7. CONCLUSION This study demonstrated that EGF may act in the early phase to promote osteoblast growth and specific marker expression rather than the late phase involving cell differentiation/mineralization.
Collapse
Affiliation(s)
- Claude Laflamme
- Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Pavillon de médecine dentaire, Université Laval, Québec, Canada G1K 7P4
| | | | | |
Collapse
|
41
|
Seib FP, Müller K, Franke M, Grimmer M, Bornhäuser M, Werner C. Engineered Extracellular Matrices Modulate the Expression Profile and Feeder Properties of Bone Marrow-Derived Human Multipotent Mesenchymal Stromal Cells. Tissue Eng Part A 2009; 15:3161-71. [DOI: 10.1089/ten.tea.2008.0600] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- F. Philipp Seib
- Leibniz Institute for Polymer Research, Max Bergmann Centre for Biomaterials Dresden, Dresden, Germany
- University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Katrin Müller
- University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Martina Franke
- Leibniz Institute for Polymer Research, Max Bergmann Centre for Biomaterials Dresden, Dresden, Germany
| | - Milauscha Grimmer
- Leibniz Institute for Polymer Research, Max Bergmann Centre for Biomaterials Dresden, Dresden, Germany
| | - Martin Bornhäuser
- University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Carsten Werner
- Leibniz Institute for Polymer Research, Max Bergmann Centre for Biomaterials Dresden, Dresden, Germany
| |
Collapse
|
42
|
The growth and differentiation of mesenchymal stem and progenitor cells cultured on aligned collagen matrices. Biomaterials 2009; 30:5950-8. [DOI: 10.1016/j.biomaterials.2009.07.039] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 07/22/2009] [Indexed: 12/31/2022]
|
43
|
Deschaseaux F, Sensébé L, Heymann D. Mechanisms of bone repair and regeneration. Trends Mol Med 2009; 15:417-29. [PMID: 19740701 DOI: 10.1016/j.molmed.2009.07.002] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 07/07/2009] [Accepted: 07/08/2009] [Indexed: 12/13/2022]
Abstract
Bone problems can have a highly deleterious impact on life and society, therefore understanding the mechanisms of bone repair is important. In vivo studies show that bone repair processes in adults resemble normal development of the skeleton during embryogenesis, which can thus be used as a model. In addition, recent studies of skeletal stem cell biology have underlined several crucial molecular and cellular processes in bone formation. Hedgehog, parathyroid hormone-related protein, Wnt, bone morphogenetic proteins and mitogen-activated protein kinases are the main molecular players, and osteoclasts and mesenchymal stem cells are the main cells involved in these processes. However, questions remain regarding the precise mechanisms of bone formation, how the different molecular processes interact, and the real identity of regenerative cells. Here, we review recent studies of bone regeneration and repair. A better understanding of the underlying mechanisms is expected to facilitate the development of new strategies for improving bone repair.
Collapse
Affiliation(s)
- Frédéric Deschaseaux
- Etablissement Français du Sang Centre-Atlantique, Groupe de Recherche sur les Cellules Souches Mésenchymateuses, Tours, France.
| | | | | |
Collapse
|