51
|
Li R, Xu J, Wong DSH, Li J, Zhao P, Bian L. Self-assembled N-cadherin mimetic peptide hydrogels promote the chondrogenesis of mesenchymal stem cells through inhibition of canonical Wnt/β-catenin signaling. Biomaterials 2017; 145:33-43. [PMID: 28843065 DOI: 10.1016/j.biomaterials.2017.08.031] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 12/14/2022]
Abstract
N-cadherin, a transmembrane protein and major component of adherens junction, mediates cell-cell interactions and intracellular signaling that are important to the regulation of cell behaviors and organ development. Previous studies have identified mimetic peptides that possess similar bioactivity as that of N-cadherin, which promotes chondrogenesis of human mesenchymal stem cells (hMSCs); however, the molecular mechanism remains unknown. In this study, we combined the N-cadherin mimetic peptide (HAVDI) with the self-assembling KLD-12 peptide: the resultant peptide is capable of self-assembling into hydrogels functionalized with N-cadherin peptide in phosphate-buffered saline (PBS) at 37 °C. Encapsulation of hMSCs in these hydrogels showed enhanced expression of chondrogenic marker genes and deposition of cartilage specific extracellular matrix rich in proteoglycan and Type II Collagen compared to control hydrogels, with a scrambled-sequence peptide after 14 days of chondrogenic culture. Furthermore, western blot showed a significantly higher expression of active glycogen synthase kinase-3β (GSK-3β), which phosphorylates β-catenin and facilitates ubiquitin-mediated degradation, as well as a lower expression of β-catenin and LEF1 in the N-cadherin peptide hydrogels versus controls. Immunofluorescence staining revealed significantly less nuclear localization of β-catenin in N-cadherin mimetic peptide hydrogels. Our findings suggest that N-cadherin peptide hydrogels suppress canonical Wnt signaling in hMSCs by reducing β-catenin nuclear translocation and the associated transcriptional activity of β-catenin/LEF-1/TCF complex, thereby enhancing the chondrogenesis of hMSCs. Our biomimetic self-assembled peptide hydrogels can serve as a tailorable and versatile three-dimensional culture platform to investigate the effect of biofunctionalization on stem cell behavior.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Jianbin Xu
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China; Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Dexter Siu Hong Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Jinming Li
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Pengchao Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong; China Orthopedic Regenerative Medicine Group (CORaMed), Hangzhou, PR China; Centre for Novel Biomaterials, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| |
Collapse
|
52
|
Lin DPL, Carnagarin R, Dharmarajan A, Dass CR. Transdifferentiation of myoblasts into osteoblasts – possible use for bone therapy. J Pharm Pharmacol 2017; 69:1661-1671. [DOI: 10.1111/jphp.12790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/18/2017] [Indexed: 02/06/2023]
Abstract
Abstract
Objectives
Transdifferentiation is defined as the conversion of one cell type to another and is an ever-expanding field with a growing number of cells found to be capable of such a process. To date, the fact remains that there are limited treatment options for fracture healing, osteoporosis and bone repair post-destruction by bone tumours. Hence, this review focuses on the transdifferentiation of myoblast to osteoblast as a means to further understand the transdifferentiation process and to investigate a potential therapeutic option if successful.
Key findings
The potent osteoinductive effects of the bone morphogenetic protein-2 are largely implicated in the transdifferentiation of myoblast to osteoblast. Bone morphogenetic protein-2-induced activation of the Smad1 protein ultimately results in JunB synthesis, the first transcriptional step in myoblast dedifferentiation. The upregulation of the activating protein-1 binding activity triggers the transcription of the runt-related transcription factor 2 gene, a transcription factor that plays a major role in osteoblast differentiation.
Summary
This potential transdifferentiation treatment may be utilised for dental implants, fracture healing, osteoporosis and bone repair post-destruction by bone tumours.
Collapse
Affiliation(s)
- Daphne P L Lin
- School of Pharmacy, Curtin University, Bentley, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA, Australia
| | - Revathy Carnagarin
- School of Pharmacy, Curtin University, Bentley, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA, Australia
| | - Arun Dharmarajan
- School of Pharmacy, Curtin University, Bentley, Perth, WA, Australia
- School of Biomedical Science, Curtin University, Bentley, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy, Curtin University, Bentley, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA, Australia
| |
Collapse
|
53
|
Cortini M, Avnet S, Baldini N. Mesenchymal stroma: Role in osteosarcoma progression. Cancer Lett 2017; 405:90-99. [PMID: 28774797 DOI: 10.1016/j.canlet.2017.07.024] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 12/21/2022]
Abstract
The initiation and progression of malignant tumors are supported by their microenvironment: cancer cells per se cannot explain growth and formation of the primary or metastasis, and a combination of proliferating tumor cells, cancer stem cells, immune cells mesenchymal stromal cells and/or cancer-associated fibroblasts all contribute to the tumor bulk. The interaction between these multiple players, under different microenvironmental conditions of biochemical and physical stimuli (i.e. oxygen tension, pH, matrix mechanics), regulates the production and biological activity of several soluble factors, extracellular matrix components, and extracellular vesicles that are needed for growth, maintenance, chemoresistance and metastatization of cancer. In osteosarcoma, a very aggressive cancer of young adults characterized by the extensive need for more effective therapies, this aspect has been only recently explored. In this view, we will discuss the role of stroma, with a particular focus on the mesenchymal stroma, contributing to osteosarcoma progression through inherent features for homing, neovascularization, paracrine cross-feeding, microvesicle secretion, and immune modulation, and also by responding to the changes of the microenvironment that are induced by tumor cells. The most recent advances in the molecular cues triggered by cytokines, soluble factors, and metabolites that are partially beginning to unravel the axis between stromal elements of mesenchymal origin and osteosarcoma cells, will be reviewed providing insights likely to be used for novel therapeutic approaches against sarcomas.
Collapse
Affiliation(s)
- Margherita Cortini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
54
|
Bermeo S, Al-Saedi A, Kassem M, Vidal C, Duque G. The Role of the Nuclear Envelope Protein MAN1 in Mesenchymal Stem Cell Differentiation. J Cell Biochem 2017; 118:4425-4435. [PMID: 28449239 DOI: 10.1002/jcb.26096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/26/2017] [Indexed: 01/09/2023]
Abstract
Mutations in MAN1, a protein of the nuclear envelope, cause bone phenotypes characterized by hyperostosis. The mechanism of this pro-osteogenic phenotype remains unknown. We increased and decreased MAN1 expression in mesenchymal stem cells (MSC) upon which standard osteogenic and adipogenic differentiation were performed. MAN1 knockdown increased osteogenesis and mineralization. In contrast, osteogenesis remained stable upon MAN1 overexpression. Regarding a mechanism, we found that low levels of MAN1 facilitated the nuclear accumulation of regulatory smads and smads-related complexes, with a concurrently high expression of nuclear β-Catenin. In addition, we found adipogenesis to be decreased in both conditions, although predominantly affected by MAN1 overexpression. Finally, lamin A, a protein of the nuclear envelope that regulates MSC differentiation, was unaffected by changes in MAN1. In conclusion, our studies demonstrated that lower levels of MAN1 in differentiating MSC are associated with higher osteogenesis and lower adipogenesis. High levels of MAN1 only affected adipogenesis. These effects could have an important role in the understanding of the role of the proteins of the nuclear envelope in bone formation. J. Cell. Biochem. 118: 4425-4435, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sandra Bermeo
- Sydney Medical School Nepean, The University of Sydney, Penrith, NSW, Australia.,Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Ahmed Al-Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, VIC, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Odense C, Denmark
| | - Christopher Vidal
- Sydney Medical School Nepean, The University of Sydney, Penrith, NSW, Australia
| | - Gustavo Duque
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, VIC, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
55
|
Kim BB, Kim M, Park YH, Ko Y, Park JB. Short-term application of dexamethasone on stem cells derived from human gingiva reduces the expression of RUNX2 and β-catenin. J Int Med Res 2017; 45:993-1006. [PMID: 28459354 PMCID: PMC5536397 DOI: 10.1177/0300060517701035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022] Open
Abstract
Objective Next-generation sequencing was performed to evaluate the effects of short-term application of dexamethasone on human gingiva-derived mesenchymal stem cells. Methods Human gingiva-derived stem cells were treated with a final concentration of 10-7 M dexamethasone and the same concentration of vehicle control. This was followed by mRNA sequencing and data analysis, gene ontology and pathway analysis, quantitative real-time polymerase chain reaction of mRNA, and western blot analysis of RUNX2 and β-catenin. Results In total, 26,364 mRNAs were differentially expressed. Comparison of the results of dexamethasone versus control at 2 hours revealed that 7 mRNAs were upregulated and 25 mRNAs were downregulated. The application of dexamethasone reduced the expression of RUNX2 and β-catenin in human gingiva-derived mesenchymal stem cells. Conclusion The effects of dexamethasone on stem cells were evaluated with mRNA sequencing, and validation of the expression was performed with qualitative real-time polymerase chain reaction and western blot analysis. The results of this study can provide new insights into the role of mRNA sequencing in maxillofacial areas.
Collapse
Affiliation(s)
- Bo-Bae Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Minji Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Youngkyung Ko
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
56
|
Li Y, Qiu SS, Shao Y, Song HH, Li GL, Lu W, Zhu LM. Dickkopf-1 has an Inhibitory Effect on Mesenchymal Stem Cells to Fibroblast Differentiation. Chin Med J (Engl) 2017; 129:1200-7. [PMID: 27174329 PMCID: PMC4878166 DOI: 10.4103/0366-6999.181974] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) are bone marrow stem cells which play an important role in tissue repair. The treatment with MSCs will be likely to aggravate the degree of fibrosis. The Wnt/β-catenin signaling pathway is involved in developmental and physiological processes, such as fibrosis. Dickkopfs (DKKs) are considered as an antagonist to block Wnt/β-catenin signaling pathway by binding the receptor of receptor-related protein (LRP5/6). DKK1 was chosen in attempt to inhibit fibrosis of MSCs by lowering activity of Wnt/β-catenin signaling pathway. Methods: Stable MSCs were randomly divided into four groups: MSCs control, MSCs + transforming growth factor-β (TGF-β), MSCs + DKK1, and MSCs + TGF-β + DKK1. Flow cytometry was used to identify MSCs. Cell viability was evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide test. Immunofluorescence was used to detect protein expression in the Wnt/β-catenin signaling pathways. Western blotting analysis was employed to test expression of fibroblast surface markers and, finally, real-time reverse transcription polymerase chain reaction was employed to test mRNA expression of fibroblast surface markers and Wnt/β-catenin signaling proteins. Results: Cultivated MSCs were found to conform to the characteristics of standard MSCs: expression of cluster of differentiation (CD) 73, 90, and 105, not expression of 34, 45, and 79. We found that DKK1 could maintain the normal cell morphology of MSCs. Western blotting analysis showed that fibroblast surface markers were expressed in high quantities in the group MSCs + TGF-β. However, the expression was lower in the MSCs + TGF-β + DKK1. Immunofluorescence showed high expression of all Wnt/β-catnin molecules in the MSCs + TGF-β group but expressed in lower quantities in MSCs + TGF-β + DKK1 group. Finally, mRNA expression of fibroblast markers vimentin, α-smooth muscle actin and Wnt/β-catenin signaling proteins β-catenin, T-cell factor, and glycogen synthase kinase-3β was significantly increased in MSCs + TGF-β group compared to control (P < 0.05). Expression of the same fibroblast markers and Wnt/β-catenin was decreased to regular quantities in the MSCs + TGF-β + DKK1 group. Conclusions: DKK1, Wnt/β-catenin inhibitors, blocks the Wnt/β-catenin signaling pathway to inhibit the process of MSCs fibrosis. It might provide some new ways for clinical treatment of certain diseases.
Collapse
Affiliation(s)
- Yan Li
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Sang-Sang Qiu
- Department of Infection Management, Affiliated Wuxi People's Hospital to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Yan Shao
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Hong-Huan Song
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Gu-Li Li
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Wei Lu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Li-Mei Zhu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| |
Collapse
|
57
|
Pigossi SC, Medeiros MC, Saska S, Cirelli JA, Scarel-Caminaga RM. Role of Osteogenic Growth Peptide (OGP) and OGP(10-14) in Bone Regeneration: A Review. Int J Mol Sci 2016; 17:ijms17111885. [PMID: 27879684 PMCID: PMC5133884 DOI: 10.3390/ijms17111885] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/24/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022] Open
Abstract
Bone regeneration is a process that involves several molecular mediators, such as growth factors, which directly affect the proliferation, migration and differentiation of bone-related cells. The osteogenic growth peptide (OGP) and its C-terminal pentapeptide OGP(10–14) have been shown to stimulate the proliferation, differentiation, alkaline phosphatase activity and matrix mineralization of osteoblastic lineage cells. However, the exact molecular mechanisms that promote osteoblastic proliferation and differentiation are not completely understood. This review presents the main chemical characteristics of OGP and/or OGP(10–14), and also discusses the potential molecular pathways induced by these growth factors to promote proliferation and differentiation of osteoblasts. Furthermore, since these peptides have been extensively investigated for bone tissue engineering, the clinical applications of these peptides for bone regeneration are discussed.
Collapse
Affiliation(s)
- Suzane C Pigossi
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP-São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
- Department of Morphology, School of Dentistry, UNESP- São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
| | - Marcell C Medeiros
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP-São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
| | - Sybele Saska
- Department of General and Inorganic Chemistry, Institute of Chemistry, UNESP-São Paulo State University, Professor Francisco Degni St, 55, CEP 14800-900 Araraquara, São Paulo, Brazil.
| | - Joni A Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP-São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
| | - Raquel M Scarel-Caminaga
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP-São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
- Department of Morphology, School of Dentistry, UNESP- São Paulo State University, Humaita St, 1680, CEP 14801-903 Araraquara, São Paulo, Brazil.
| |
Collapse
|
58
|
Wang T, Zhang X, Bikle DD. Osteogenic Differentiation of Periosteal Cells During Fracture Healing. J Cell Physiol 2016; 232:913-921. [PMID: 27731505 DOI: 10.1002/jcp.25641] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 12/15/2022]
Abstract
Five to ten percent of fractures fail to heal normally leading to additional surgery, morbidity, and altered quality of life. Fracture healing involves the coordinated action of stem cells primarily coming from the periosteum which differentiate into the chondrocytes and osteoblasts, forming first the soft (cartilage) callus followed by the hard (bone) callus. These stem cells are accompanied by a vascular invasion that appears critical for the differentiation process and which may enable the entry of osteoclasts necessary for the remodeling of the callus into mature bone. However, more research is needed to clarify the signaling events that activate the osteochondroprogenitor cells of periosteum and stimulate their differentiation into chondrocytes and osteoblasts. Ultimately a thorough understanding of the mechanisms for differential regulation of these osteochondroprogenitors will aid in the treatment of bone healing and the prevention of delayed union and nonunion of fractures. In this review, evidence supporting the concept that the periosteal cells are the major cell sources of skeletal progenitors for the fracture callus will be discussed. The osteogenic differentiation of periosteal cells manipulated by Wnt/β-catenin, TGF/BMP, Ihh/PTHrP, and IGF-1/PI3K-Akt signaling in fracture repair will be examined. The effect of physical (hypoxia and hyperoxia) and chemical factors (reactive oxygen species) as well as the potential coordinated regulatory mechanisms in the periosteal progenitor cells promoting osteogenic differentiation will also be discussed. Understanding the regulation of periosteal osteochondroprogenitors during fracture healing could provide insight into possible therapeutic targets and thereby help to enhance future fracture healing and bone tissue engineering approaches. J. Cell. Physiol. 232: 913-921, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tao Wang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York.,Endocrine Unit, VA Medical Center and University of California, San Francisco, California
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Daniel D Bikle
- Endocrine Unit, VA Medical Center and University of California, San Francisco, California
| |
Collapse
|
59
|
Liu S, Sun Y, Fu Y, Chang D, Fu C, Wang G, Liu Y, Tay FR, Zhou Y. Bioinspired Collagen-Apatite Nanocomposites for Bone Regeneration. J Endod 2016; 42:1226-32. [PMID: 27377439 DOI: 10.1016/j.joen.2016.04.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Natural bone has a complex hierarchical nanostructure composed of well-organized collagen fibrils embedded with apatite crystallites. Bone tissue engineering requires scaffolds with structural properties and functionality similar to the natural bone. Inspired by bone, a collagen-apatite (Col-Ap) nanocomposite was fabricated with bonelike subfibrillar nanostructures using a modified bottom-up biomimetic approach and has a potential role in the healing of large bone defects in unresolved apical periodontitis. METHODS The bone regeneration potential of the Col-Ap nanocomposite was investigated by comparing it with inorganic beta-tricalcium phosphate and organic pure collagen using a critical-sized rodent mandibular defect model. Micro-computed tomographic imaging and histologic staining were used to evaluate new bone formation in vivo. RESULTS When compared with the beta-tricalcium phosphate and collagen scaffolds, the Col-Ap nanocomposite scaffold exhibited superior regeneration properties characterized by profuse deposition of new bony structures and vascularization at the defect center. Immunohistochemistry showed that the transcription factor osterix and vascular endothelial growth factor receptor 1 were highly expressed in the Col-Ap group. The results indicate that the Col-Ap nanocomposite activates more bone-forming cells and stimulates more vascular tissue ingrowth. Furthermore, the Col-Ap nanocomposite induces extracellular matrix secretion and mineralization of rat bone marrow stem cells. The increased expression of transforming growth factor beta 1 may contribute to the formation of a mineralized extracellular matrix. CONCLUSIONS The present study lays the foundation for the development of Col-Ap nanocomposite-based bone grafts for future clinical applications in bone regeneration of large periapical lesions after apical curettage or apicoectomy.
Collapse
Affiliation(s)
- Shuai Liu
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yue Sun
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yu Fu
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Datong Chang
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Cuicui Fu
- Department of Orthodontics, School of Stomatology, Zhengzhou University, Henan, People's Republic of China
| | - Gaonan Wang
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yan Liu
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.
| | - Franklin R Tay
- Department of Endodontics, The Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Yanheng Zhou
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.
| |
Collapse
|
60
|
Liu YK, Zhou ZY, Liu F. Transcriptome changes during TNF-α promoted osteogenic differentiation of dental pulp stem cells (DPSCs). Biochem Biophys Res Commun 2016; 476:426-430. [DOI: 10.1016/j.bbrc.2016.05.137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 05/25/2016] [Indexed: 01/09/2023]
|
61
|
Canonical Wnt/β-Catenin Signaling Pathway Is Dysregulated in Patients With Primary and Secondary Myelofibrosis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:523-526. [PMID: 27381374 DOI: 10.1016/j.clml.2016.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/12/2016] [Accepted: 06/01/2016] [Indexed: 11/21/2022]
Abstract
INTRODUCTION β-Catenin is a central effector molecule of the canonical wingless-related integration site (Wnt) signaling pathway. It is important for maintenance of stem cell homeostasis and its aberrant activation has been implicated in a wide array of malignant hematological disorders. There are few reports suggesting its dysregulation in Philadelphia chromosome-negative (Ph-) myeloproliferative neoplasms (MPNs). PATIENTS AND METHODS We analyzed β-catenin mRNA expression in bone marrow (BM) aspirates of 29 patients with primary (PMF) and 4 patients with secondary, post Ph- MPN, myelofibrosis (SMF) using quantitative real-time polymerase chain reaction (qRT PCR). The control group consisted of 16 BM aspirates from patients with limited-stage aggressive non-Hodgkin lymphoma without BM involvement. We compared relative gene expression with clinical and hematological parameters. RESULTS Relative expression of β-catenin differed significantly among groups (P = .0002), it was significantly higher in patients with PMF and SMF than in the control group, but did not differ between patients with PMF and SMF. A negative correlation was found regarding hemoglobin level in PMF (P = .017). No association according to Janus kinase 2 (JAK2) V617F mutational status or JAK2 V617F allele burden was detected. CONCLUSION Our results show for the first time that β-catenin mRNA expression is increased in patients with PMF and SMF and its upregulation might potentiate anemia. A number of inflammatory cytokines associated with PMF are capable of mediating their effects through increased β-catenin expression. Accordingly, β-catenin can induce expression of a number of genes implicated in processes of cell cycle control, fibrosis, and angiogenesis, which are central to the PMF pathogenesis. Therefore, β-catenin might represent an interesting new therapeutic target in these diseases.
Collapse
|
62
|
A Nexus Consisting of Beta-Catenin and Stat3 Attenuates BRAF Inhibitor Efficacy and Mediates Acquired Resistance to Vemurafenib. EBioMedicine 2016; 8:132-149. [PMID: 27428425 PMCID: PMC4919613 DOI: 10.1016/j.ebiom.2016.04.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 02/06/2023] Open
Abstract
Acquired resistance to second generation BRAF inhibitors (BRAFis), like vemurafenib is limiting the benefits of long term targeted therapy for patients with malignant melanomas that harbor BRAF V600 mutations. Since many resistance mechanisms have been described, most of them causing a hyperactivation of the MAPK- or PI3K/AKT signaling pathways, one potential strategy to overcome BRAFi resistance in melanoma cells would be to target important common signaling nodes. Known factors that cause secondary resistance include the overexpression of receptor tyrosine kinases (RTKs), alternative splicing of BRAF or the occurrence of novel mutations in MEK1 or NRAS. In this study we show that β-catenin is stabilized and translocated to the nucleus in approximately half of the melanomas that were analyzed and which developed secondary resistance towards BRAFi. We further demonstrate that β-catenin is involved in the mediation of resistance towards vemurafenib in vitro and in vivo. Unexpectedly, β-catenin acts mainly independent of the TCF/LEF dependent canonical Wnt-signaling pathway in resistance development, which partly explains previous contradictory results about the role of β-catenin in melanoma progression and therapy resistance. We further demonstrate that β-catenin interacts with Stat3 after chronic vemurafenib treatment and both together cooperate in the acquisition and maintenance of resistance towards BRAFi.
Collapse
|
63
|
Wu M, Chen G, Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res 2016; 4:16009. [PMID: 27563484 PMCID: PMC4985055 DOI: 10.1038/boneres.2016.9] [Citation(s) in RCA: 1146] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) and bone morphogenic protein (BMP) signaling has fundamental roles in both embryonic skeletal development and postnatal bone homeostasis. TGF-βs and BMPs, acting on a tetrameric receptor complex, transduce signals to both the canonical Smad-dependent signaling pathway (that is, TGF-β/BMP ligands, receptors, and Smads) and the non-canonical-Smad-independent signaling pathway (that is, p38 mitogen-activated protein kinase/p38 MAPK) to regulate mesenchymal stem cell differentiation during skeletal development, bone formation and bone homeostasis. Both the Smad and p38 MAPK signaling pathways converge at transcription factors, for example, Runx2 to promote osteoblast differentiation and chondrocyte differentiation from mesenchymal precursor cells. TGF-β and BMP signaling is controlled by multiple factors, including the ubiquitin–proteasome system, epigenetic factors, and microRNA. Dysregulated TGF-β and BMP signaling result in a number of bone disorders in humans. Knockout or mutation of TGF-β and BMP signaling-related genes in mice leads to bone abnormalities of varying severity, which enable a better understanding of TGF-β/BMP signaling in bone and the signaling networks underlying osteoblast differentiation and bone formation. There is also crosstalk between TGF-β/BMP signaling and several critical cytokines’ signaling pathways (for example, Wnt, Hedgehog, Notch, PTHrP, and FGF) to coordinate osteogenesis, skeletal development, and bone homeostasis. This review summarizes the recent advances in our understanding of TGF-β/BMP signaling in osteoblast differentiation, chondrocyte differentiation, skeletal development, cartilage formation, bone formation, bone homeostasis, and related human bone diseases caused by the disruption of TGF-β/BMP signaling.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| | - Guiqian Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA; Department of neurology, Bruke Medical Research Institute, Weil Cornell Medicine of Cornell University, White Plains, USA
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| |
Collapse
|
64
|
Zhou S, Thornhill TS, Meng F, Xie L, Wright J, Glowacki J. Influence of osteoarthritis grade on molecular signature of human cartilage. J Orthop Res 2016; 34:454-62. [PMID: 26336057 DOI: 10.1002/jor.23043] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 02/04/2023]
Abstract
Articular chondrocytes maintain cartilage matrix turnover and have the capacity for anabolic and catabolic activities that can be influenced by injury and disease. This study tested the hypothesis that catabolic genes are upregulated with regional osteoarthritis (OA) disease severity within a joint. With IRB approval, specimens of knee cartilage obtained as discarded tissues from subjects undergoing arthroplasty were partitioned for each subject by OA disease severity and evaluated for gene expression by RT-PCR. There was regional OA grade-associated upregulation of expected inflammatory mediators TNF-α, TNF receptors, IFN-γ, and interleukins as well as genes encoding proteolytic enzymes, including Adamts-5 and MMPs. Osteoclast-related genes, cathepsin K, tartrate-resistant acid phosphatase (TRAP), RANKL, RANK, M-CSF, and c-fms, but not osteoprotegerin, were induced in advanced grades. In vitro treatment of normal human chondrocytes with interleukin-1β upregulated similar genes; this provides evidence that chondrocytes per se can be the source of osteoclast-related factors. Immunohistochemical staining showed that RANK- and RANKL-positive cells were abundant in advanced grades, especially in chondrocyte clusters. This suggests a possible autocrine mechanism by which an osteoclast phenotype is induced in articular chondrocytes. In sum, these studies identified gene expression signatures in human OA cartilage based upon regional disease severity within a joint. There was an effect of OA Grade on expression of osteoclastic lytic enzymes and regulatory factors in human articular chondrocytes. Induction of an osteoclast-like phenotype in chondrocytes may be part of OA progression and suggests specific therapeutic approaches.
Collapse
Affiliation(s)
- Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Thomas S Thornhill
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fangang Meng
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Xie
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - John Wright
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Oral and Maxillofacial Surgery, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
65
|
Spanjer AIR, Baarsma HA, Oostenbrink LM, Jansen SR, Kuipers CC, Lindner M, Postma DS, Meurs H, Heijink IH, Gosens R, Königshoff M. TGF-β-induced profibrotic signaling is regulated in part by the WNT receptor Frizzled-8. FASEB J 2016; 30:1823-35. [PMID: 26849959 DOI: 10.1096/fj.201500129] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
Abstract
TGF-β is important in lung injury and remodeling processes. TGF-β and Wingless/integrase-1 (WNT) signaling are interconnected; however, the WNT ligand-receptor complexes involved are unknown. Thus, we aimed to identify Frizzled (FZD) receptors that mediate TGF-β-induced profibrotic signaling. MRC-5 and primary human lung fibroblasts were stimulated with TGF-β1, WNT-5A, or WNT-5B in the presence and absence of specific pathway inhibitors. Specific small interfering RNA was used to knock down FZD8. In vivo studies using bleomycin-induced lung fibrosis were performed in wild-type and FZD8-deficient mice. TGF-β1 induced FZD8 specifically via Smad3-dependent signaling in MRC-5 and primary human lung fibroblasts. It is noteworthy that FZD8 knockdown reduced TGF-β1-induced collagen Iα1, fibronectin, versican, α-smooth muscle (sm)-actin, and connective tissue growth factor. Moreover, bleomycin-induced lung fibrosis was attenuated in FZD8-deficient mice in vivo Although inhibition of canonical WNT signaling did not affect TGF-β1-induced gene expression in vitro, noncanonical WNT-5B mimicked TGF-β1-induced fibroblast activation. FZD8 knockdown reduced both WNT-5B-induced gene expression of fibronectin and α-sm-actin, as well as WNT-5B-induced changes in cellular impedance. Collectively, our findings demonstrate a role for FZD8 in TGF-β-induced profibrotic signaling and imply that WNT-5B may be the ligand for FZD8 in these responses.-Spanjer, A. I. R., Baarsma, H. A., Oostenbrink, L. M., Jansen, S. R., Kuipers, C. C., Lindner, M., Postma, D. S., Meurs, H., Heijink, I. H., Gosens, R., Königshoff, M. TGF-β-induced profibrotic signaling is regulated in part by the WNT receptor Frizzled-8.
Collapse
Affiliation(s)
- Anita I R Spanjer
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, German Center for Lung Research (DZL), University Hospital Grosshadern, Ludwig Maximilians University Munich, Munich, Germany
| | - Lisette M Oostenbrink
- Comprehensive Pneumology Center, Helmholtz Center Munich, German Center for Lung Research (DZL), University Hospital Grosshadern, Ludwig Maximilians University Munich, Munich, Germany
| | - Sepp R Jansen
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Christine C Kuipers
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michael Lindner
- Asklepios Fachkliniken München-Gauting, Munich, Germany; and
| | - Dirkje S Postma
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands Department of Pulmonology
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands Department of Pulmonology, Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, and
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, German Center for Lung Research (DZL), University Hospital Grosshadern, Ludwig Maximilians University Munich, Munich, Germany;
| |
Collapse
|
66
|
Liang X, Glowacki J, Hahne J, Xie L, LeBoff MS, Zhou S. Dehydroepiandrosterone Stimulation of Osteoblastogenesis in Human MSCs Requires IGF-I Signaling. J Cell Biochem 2016; 117:1769-74. [DOI: 10.1002/jcb.25475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Xiaonan Liang
- Department of Orthopedic Surgery; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
- Department of Orthopedic Trauma and Hand Surgery; The First Affiliated Hospital of Guangxi Medical University; Nanning Guangxi China
| | - Julie Glowacki
- Department of Orthopedic Surgery; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
| | - Jochen Hahne
- Department of Orthopedic Surgery; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
- MW Center for Orthopedics and Sports Medicine; Munich Germany
| | - Li Xie
- Department of Orthopedic Surgery; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
- Department of Clinical Laboratory; The First Affiliated Hospital of Guangxi Medical University; Nanning Guangxi China
| | - Meryl S. LeBoff
- Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
| | - Shuanhu Zhou
- Department of Orthopedic Surgery; Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts
- Harvard Stem Cell Institute; Harvard University; Cambridge Massachusetts
| |
Collapse
|
67
|
Liu H, Hao W, Wang X, Su H. miR-23b targets Smad 3 and ameliorates the LPS-inhibited osteogenic differentiation in preosteoblast MC3T3-E1 cells. J Toxicol Sci 2016; 41:185-93. [PMID: 26961602 DOI: 10.2131/jts.41.185] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hongzhi Liu
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Wei Hao
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Xin Wang
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Hao Su
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| |
Collapse
|
68
|
Angbohang A, Wu N, Charalambous T, Eastlake K, Lei Y, Kim YS, Sun XH, Limb GA. Downregulation of the Canonical WNT Signaling Pathway by TGFβ1 Inhibits Photoreceptor Differentiation of Adult Human Müller Glia with Stem Cell Characteristics. Stem Cells Dev 2016; 25:1-12. [PMID: 26456050 PMCID: PMC4692127 DOI: 10.1089/scd.2015.0262] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/09/2015] [Indexed: 12/29/2022] Open
Abstract
Müller glia are responsible for the retina regeneration observed in zebrafish. Although the human retina harbors Müller glia with stem cell characteristics, there is no evidence that they regenerate the retina after disease or injury. Transforming growth factor-β (TGFβ) and Wnt signaling regulate retinal neurogenesis and inflammation, but their roles in the neural differentiation of human Müller stem cells (hMSC) are not known. We examined hMSC lines in vitro for the expression of various Wnt signaling components and for their modulation by TGFβ1, as well as the effect of this cytokine on the photoreceptor differentiation of these cells. Culture of hMSC with a combination of factors that induce photoreceptor differentiation of hMSC (FGF2, taurine, retinoic acid, and insulin-like growth factor type1; FTRI), markedly upregulated the expression of components of the canonical Wnt signaling pathway, including WNT2B, DKK1, and active β-CATENIN. Although FTRI did not modify mRNA expression of WNT5B, a component of the noncanonical/planar cell polarity Wnt pathway, it upregulated its secretion. Furthermore, TGFβ1 not only decreased WNT2B expression, but also inhibited FTRI-induced photoreceptor differentiation of hMSC, as determined by expression of the photoreceptor markers NR2E3, RHODOPSIN, and RECOVERIN. Inhibition of TGFβ1 signaling by an ALK5 inhibitor prevented TGFβ1-induced changes in the expression of the two Wnt ligands examined. More importantly, inhibition of the canonical WNT signaling by XAV-939 prevented FTRI-induced photoreceptor differentiation. These observations suggest that TGFβ may play a key role in preventing neural differentiation of hMSC and may constitute a potential target for induction of endogenous regeneration of the human retina.
Collapse
Affiliation(s)
- Angshumonik Angbohang
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Na Wu
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Thalis Charalambous
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Karen Eastlake
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Yuan Lei
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yung Su Kim
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Xinghuai H. Sun
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
69
|
Biressi S, Miyabara EH, Gopinath SD, Carlig PMM, Rando TA. A Wnt-TGFβ2 axis induces a fibrogenic program in muscle stem cells from dystrophic mice. Sci Transl Med 2015; 6:267ra176. [PMID: 25520397 DOI: 10.1126/scitranslmed.3008411] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have previously observed that Wnt signaling activates a fibrogenic program in adult muscle stem cells, called satellite cells, during aging. We genetically labeled satellite cells in a mouse model of Duchenne muscular dystrophy to follow their fate during the progression of the disease. We observed that a fraction of satellite cells had a reduced myogenic potential and showed enhanced expression of profibrotic genes compared to age-matched controls. By combining in vitro and in vivo results, we found that expression of transforming growth factor-β2 (TGFβ2) was induced in response to elevated canonical Wnt signaling in dystrophic muscles and that the resulting increase in TGFβ activity affected the behavior of satellite cells in an autocrine or paracrine fashion. Indeed, pharmacological inhibition of the TGFβ pathway in vivo reduced the fibrogenic characteristics of satellite cells. These studies shed new light on the cellular and molecular mechanisms responsible for stem cell dysfunction in dystrophic muscle and may contribute to the development of more effective and specific therapeutic approaches for the prevention of muscle fibrosis.
Collapse
Affiliation(s)
- Stefano Biressi
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elen H Miyabara
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA. Anatomy Department, Institute of Biomedical Sciences, University of São Paulo, 2415 Lineu Prestes Avenue, São Paulo, São Paulo 05508-000, Brazil
| | - Suchitra D Gopinath
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Poppy M M Carlig
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas A Rando
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA. Neurology Service, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA.
| |
Collapse
|
70
|
Suen PK, Qin L. Sclerostin, an emerging therapeutic target for treating osteoporosis and osteoporotic fracture: A general review. J Orthop Translat 2015; 4:1-13. [PMID: 30035061 PMCID: PMC5987014 DOI: 10.1016/j.jot.2015.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/02/2015] [Accepted: 08/12/2015] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis and its associated fracture risk has become one of the major health burdens in our aging population. Currently, bisphosphonate, one of the most popular antiresorptive drugs, is used widely to treat osteoporosis but so far still no consensus has been reached for its application in treatment of osteoporotic fractures. However, in old patients, boosting new bone formation and its remodelling is essential for bone healing in age-related osteoporosis and osteoporotic fractures. Sclerostin, an inhibitor of the Wnt/β-catenin signalling pathway that regulates bone growth, has become an attractive therapeutic target for treating osteoporosis. In this review, we summarize the recent findings of sclerostin and its potential as an effective drug target for treating both osteoporosis and osteoporotic fractures.
Collapse
Affiliation(s)
- Pui Kit Suen
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
71
|
Singh SP, Tao S, Fields TA, Webb S, Harris RC, Rao R. Glycogen synthase kinase-3 inhibition attenuates fibroblast activation and development of fibrosis following renal ischemia-reperfusion in mice. Dis Model Mech 2015; 8:931-40. [PMID: 26092126 PMCID: PMC4527294 DOI: 10.1242/dmm.020511] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/26/2015] [Indexed: 01/06/2023] Open
Abstract
Glycogen synthase kinase-3β (GSK3β) is a serine/threonine protein kinase that plays an important role in renal tubular injury and regeneration in acute kidney injury. However, its role in the development of renal fibrosis, often a long-term consequence of acute kidney injury, is unknown. Using a mouse model of renal fibrosis induced by ischemia-reperfusion injury, we demonstrate increased GSK3β expression and activity in fibrotic kidneys, and its presence in myofibroblasts in addition to tubular epithelial cells. Pharmacological inhibition of GSK3 using TDZD-8 starting before or after ischemia-reperfusion significantly suppressed renal fibrosis by reducing the myofibroblast population, collagen-1 and fibronectin deposition, inflammatory cytokines, and macrophage infiltration. GSK3 inhibition in vivo reduced TGF-β1, SMAD3 activation and plasminogen activator inhibitor-1 levels. Consistently in vitro, TGF-β1 treatment increased GSK3β expression and GSK3 inhibition abolished TGF-β1-induced SMAD3 activation and α-smooth muscle actin (α-SMA) expression in cultured renal fibroblasts. Importantly, overexpression of constitutively active GSK3β stimulated α-SMA expression even in the absence of TGF-β1 treatment. These results suggest that TGF-β regulates GSK3β, which in turn is important for TGF-β–SMAD3 signaling and fibroblast-to-myofibroblast differentiation. Overall, these studies demonstrate that GSK3 could promote renal fibrosis by activation of TGF-β signaling and the use of GSK3 inhibitors might represent a novel therapeutic approach for progressive renal fibrosis that develops as a consequence of acute kidney injury. Summary: GSK3 promotes renal fibrosis by activation of TGF-β signaling, and the use of GSK3 inhibitors might represent a novel therapeutic approach for progressive renal fibrosis that develops as a consequence of acute kidney injury.
Collapse
Affiliation(s)
- Shailendra P Singh
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Shixin Tao
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Timothy A Fields
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Sydney Webb
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Reena Rao
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| |
Collapse
|
72
|
Zhou S. Paracrine effects of haematopoietic cells on human mesenchymal stem cells. Sci Rep 2015; 5:10573. [PMID: 26030407 PMCID: PMC4450757 DOI: 10.1038/srep10573] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/17/2015] [Indexed: 12/28/2022] Open
Abstract
Stem cell function decline during ageing can involve both cell intrinsic and extrinsic mechanisms. Bone and blood formation are intertwined in bone marrow, therefore haematopoietic cells and bone cells could be extrinsic factors for each other. In this study, we assessed the paracrine effects of extrinsic factors from haematopoietic cells on human mesenchymal stem cells (MSCs). Our data showed that haematopoietic cells stimulate proliferation, osteoblast differentiation and inhibit senescence of MSCs; TNF-α, PDGF-β, Wnt1, 4, 6, 7a and 10a, sFRP-3 and sFRP-5 are dominantly expressed in haematopoietic cells; the age-related increase of TNF-α in haematopoietic cells may perform as a negative factor in the interactions of haematopoietic cells on MSCs via TNF-α receptors and then activating NF-κB signaling or Wnt/β-catenin signaling to induce senescence and reduce osteoblast differentiation in MSCs. In conclusion, our data demonstrated that there are paracrine interactions of haematopoietic cells on human MSCs; immunosenescence may be one of the extrinsic mechanisms by which skeletal stem cell function decline during human skeletal ageing.
Collapse
Affiliation(s)
- Shuanhu Zhou
- 1] Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA [2] Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
73
|
Li T, Li H, Li T, Fan J, Zhao RC, Weng X. MicroRNA expression profile of dexamethasone-induced human bone marrow-derived mesenchymal stem cells during osteogenic differentiation. J Cell Biochem 2015; 115:1683-91. [PMID: 24802236 DOI: 10.1002/jcb.24831] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/28/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023]
Abstract
MiRNAs have been identified in various plants and animals where they function in post-transcriptional regulation. Although studies revealed that dexamethasone play a pivotal role in the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs), the identification of specific miRNAs and their regulatory roles in this process remain poorly defined. In this study, microarrays were used to analyze the miRNA expression profile of dexamethasone-induced hBMSCs derived from three donors, and RT-PCRs were used to confirm the microarray results. Nine upregulated miRNAs and seven downregulated miRNAs were identified. The putative target genes of these miRNAs were predicted using bioinformatics analysis. Subsequently, we focused our attention on the functional analysis of an upregulated miRNA, miR-23a. Overexpression of miR-23a inhibited osteogenic differentiation of hBMSCs at the cellular, mRNA, and protein levels. The results of our study provide an experimental basis for further research on miRNAs functions during osteogenic differentiation of dexamethasone-induced hBMSCs.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, 100730, China
| | | | | | | | | | | |
Collapse
|
74
|
Peng X, Luo Z, Kang Q, Deng D, Wang Q, Peng H, Wang S, Wei Z. FOXQ1 mediates the crosstalk between TGF-β and Wnt signaling pathways in the progression of colorectal cancer. Cancer Biol Ther 2015; 16:1099-109. [PMID: 25955104 DOI: 10.1080/15384047.2015.1047568] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A wide variety of signaling transduction pathways contribute to tumorigenesis. Forkhead box Q1 (FOXQ1) is a member of the forkhead transcription factor family and its upregulation is closely correlated with tumor progression and prognosis of multiple cancer types, including colorectal cancer. However, the molecular mechanisms by which FOXQ1 promotes tumorigenesis, especially cancer cell invasion and metastasis in colorectal cancer, have not been fully elucidated. In the present study, we demonstrate that FOXQ1 is overexpressed in colorectal tumor tissues and its expression level is closely correlated with the stage and lymph node metastasis of colorectal cancer. In in vitro cultured SW480 colorectal cancer cells, knockdown of FOXQ1 expression by small interfering RNA greatly diminished the aggressive tumor behaviors of SW480 cells, including angiogenesis, invasion, epithelial-mesenchymal transition, and resistance to chemotherapy drug-induced apoptosis. Further mechanistic investigation showed that FOXQ1 silencing prevents the nuclear translocation of β-catenin, thus reducing the activity of Wnt signaling. Moreover, TGF-β1 induced the expression of FOXQ1 as well as the migration and invasion of SW480 cells, which was partially prevented following knockdown of FOXQ1. Our results demonstrate that FOXQ1 plays a critical role during the tumorigenesis of colorectal cancer and is a mediator of the crosstalk between Wnt and TGF-β signaling pathways. Our findings provide further insight into the cancer biology of colorectal cancer and suggest that FOXQ1 is a potential therapeutic target for the development of therapies for colorectal cancer.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- 7-AAD, 7-aminoactinomycin D
- DAPI, 4′,6-diamidino-2-phenylindole
- DEPC, Diethy pyrocarbonate
- DMSO, Dimethyl sulfoxide
- EMT, Epithelial-Mesenchymal transition
- FOXQ1
- FOXQ1, Forkhead Box Q1
- L-OHP, Oxaliplatin
- MMP2, Matrix metalloproteinase-2
- MiRNA, MicroRNA
- NC-shRNA, Negative Control-shRNA
- PBS, Phosphate buffer solution
- PBS, phosphate buffered saline
- PKA, proteinkinase A
- PVDF, Polyvinylidene fluoride
- RNAi, RNA interference
- SDS, Sodium dodecyl sulfonate
- TBS, Tris-buffered saline
- TEMED, Tetra methyl ethylene diamine
- TGF-β, Transformin growth β
- TGF-β1
- Tris, Trihydroxymethyl minomethane
- VEGF-A, Vascular endothelial growth factor-A
- Wnt signaling
- aggressive tumor behavior
- cDNA, Complementary DNA
- colorectal cancer
- ddH2O, double distilled H2O
- epithelial-mesenchymal transition
- qRT-PCR, Quantitative real-time PCR
- shRNA, Short hairpin RNA
- μg, Microgramme
- μl, Microliter
Collapse
Affiliation(s)
- Xudong Peng
- a Gastrointestinal Surgical Unit; The First Affiliated Hospital of Chongqing Medical University ; Chongqing , China
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
Stem cells are cells specialized cell, capable of renewing themselves through cell division and can differentiate into multi-lineage cells. These cells are categorized as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. Mesenchymal stem cells (MSCs) are adult stem cells which can be isolated from human and animal sources. Human MSCs (hMSCs) are the non-haematopoietic, multipotent stem cells with the capacity to differentiate into mesodermal lineage such as osteocytes, adipocytes and chondrocytes as well ectodermal (neurocytes) and endodermal lineages (hepatocytes). MSCs express cell surface markers like cluster of differentiation (CD)29, CD44, CD73, CD90, CD105 and lack the expression of CD14, CD34, CD45 and HLA (human leucocyte antigen)-DR. hMSCs for the first time were reported in the bone marrow and till now they have been isolated from various tissues, including adipose tissue, amniotic fluid, endometrium, dental tissues, umbilical cord and Wharton's jelly which harbours potential MSCs. hMSCs have been cultured long-term in specific media without any severe abnormalities. Furthermore, MSCs have immunomodulatory features, secrete cytokines and immune-receptors which regulate the microenvironment in the host tissue. Multilineage potential, immunomodulation and secretion of anti-inflammatory molecules makes MSCs an effective tool in the treatment of chronic diseases. In the present review, we have highlighted recent research findings in the area of hMSCs sources, expression of cell surface markers, long-term in vitro culturing, in vitro differentiation potential, immunomodulatory features, its homing capacity, banking and cryopreservation, its application in the treatment of chronic diseases and its use in clinical trials.
Collapse
|
76
|
Khanghahi AM, Zeynali B, Akhlaghpoor A, Tafreshi AP, Krieglstein K. Activation of TGFβ1 signaling enhances early dopaminergic differentiation in unrestricted somatic stem cells. Neurosci Lett 2014; 583:60-4. [DOI: 10.1016/j.neulet.2014.08.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 07/23/2014] [Accepted: 08/05/2014] [Indexed: 01/09/2023]
|
77
|
Huang X, Xu J, Huang M, Li J, Dai L, Dai K, Zhang X. Histone deacetylase1 promotes TGF-β1-mediated early chondrogenesis through down-regulating canonical Wnt signaling. Biochem Biophys Res Commun 2014; 453:810-6. [DOI: 10.1016/j.bbrc.2014.10.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/07/2014] [Indexed: 12/22/2022]
|
78
|
de Moraes M, da Rocha Neto PC, de Matos FR, Lopes MLDDS, de Azevedo PRM, Costa ADLL. Immunoexpression of transforming growth factor beta and interferon gamma in radicular and dentigerous cysts. J Endod 2014; 40:1293-7. [PMID: 25043252 DOI: 10.1016/j.joen.2014.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 10/29/2013] [Accepted: 01/07/2014] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The aim of this study was to evaluate and compare the immunohistochemical expression of transforming growing factor beta (TGF-β) and interferon gamma (IFN-γ) between radicular cysts (RCs) and dentigerous cysts (DCs). METHODS Twenty RCs and DCs were selected for analysis of the immunoexpression of TGF-β and IFN-γ in the epithelium and capsule. RESULTS The cell reactivity of TGF-β and IFN-γ in the lining epithelium and capsule of RCs showed no significant differences when compared with DCs (P > .05). There was a tendency of a higher expression of TGF-β in the capsule of DCs. CONCLUSIONS Our results showed the presence of TGF-β and IFN-γ in RCs and DCs, supporting the hypothesis that both participate in the development of these lesions, where IFN-γ usually plays a role in bone resorption, which is counterbalanced by the osteoprotective activity performed by TGF-β.
Collapse
Affiliation(s)
- Maiara de Moraes
- Postgraduate Program, Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Pedro Carlos da Rocha Neto
- Postgraduate Program, Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Felipe Rodrigues de Matos
- Postgraduate Program, Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | | | - Antonio de Lisboa Lopes Costa
- Postgraduate Program, Oral Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
79
|
Cheung KSC, Sposito N, Stumpf PS, Wilson DI, Sanchez-Elsner T, Oreffo ROC. MicroRNA-146a regulates human foetal femur derived skeletal stem cell differentiation by down-regulating SMAD2 and SMAD3. PLoS One 2014; 9:e98063. [PMID: 24892945 PMCID: PMC4043645 DOI: 10.1371/journal.pone.0098063] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/27/2014] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRs) play a pivotal role in a variety of biological processes including stem cell differentiation and function. Human foetal femur derived skeletal stem cells (SSCs) display enhanced proliferation and multipotential capacity indicating excellent potential as candidates for tissue engineering applications. This study has examined the expression and role of miRs in human foetal femur derived SSC differentiation along chondrogenic and osteogenic lineages. Cells isolated from the epiphyseal region of the foetal femur expressed higher levels of genes associated with chondrogenesis while cells from the foetal femur diaphyseal region expressed higher levels of genes associated with osteogenic differentiation. In addition to the difference in osteogenic and chondrogenic gene expression, epiphyseal and diaphyseal cells displayed distinct miRs expression profiles. miR-146a was found to be expressed by human foetal femur diaphyseal cells at a significantly enhanced level compared to epiphyseal populations and was predicted to target various components of the TGF-β pathway. Examination of miR-146a function in foetal femur cells confirmed regulation of protein translation of SMAD2 and SMAD3, important TGF-β and activin ligands signal transducers following transient overexpression in epiphyseal cells. The down-regulation of SMAD2 and SMAD3 following overexpression of miR-146a resulted in an up-regulation of the osteogenesis related gene RUNX2 and down-regulation of the chondrogenesis related gene SOX9. The current findings indicate miR-146a plays an important role in skeletogenesis through attenuation of SMAD2 and SMAD3 function and provide further insight into the role of miRs in human skeletal stem cell differentiation modulation with implications therein for bone reparation.
Collapse
Affiliation(s)
- Kelvin S. C. Cheung
- Bone and Joint Research Group, Institute of Developmental Sciences, Southampton General Hospital, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, Southampton, United Kingdom
| | - Nunzia Sposito
- Bone and Joint Research Group, Institute of Developmental Sciences, Southampton General Hospital, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, Southampton, United Kingdom
| | - Patrick S. Stumpf
- Bone and Joint Research Group, Institute of Developmental Sciences, Southampton General Hospital, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, Southampton, United Kingdom
| | - David I. Wilson
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, Southampton, United Kingdom
| | - Tilman Sanchez-Elsner
- Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Institute of Developmental Sciences, Southampton General Hospital, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
80
|
Cartilage tissue engineering: molecular control of chondrocyte differentiation for proper cartilage matrix reconstruction. Biochim Biophys Acta Gen Subj 2014; 1840:2414-40. [PMID: 24608030 DOI: 10.1016/j.bbagen.2014.02.030] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/06/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Articular cartilage defects are a veritable therapeutic problem because therapeutic options are very scarce. Due to the poor self-regeneration capacity of cartilage, minor cartilage defects often lead to osteoarthritis. Several surgical strategies have been developed to repair damaged cartilage. Autologous chondrocyte implantation (ACI) gives encouraging results, but this cell-based therapy involves a step of chondrocyte expansion in a monolayer, which results in the loss in the differentiated phenotype. Thus, despite improvement in the quality of life for patients, reconstructed cartilage is in fact fibrocartilage. Successful ACI, according to the particular physiology of chondrocytes in vitro, requires active and phenotypically stabilized chondrocytes. SCOPE OF REVIEW This review describes the unique physiology of cartilage, with the factors involved in its formation, stabilization and degradation. Then, we focus on some of the most recent advances in cell therapy and tissue engineering that open up interesting perspectives for maintaining or obtaining the chondrogenic character of cells in order to treat cartilage lesions. MAJOR CONCLUSIONS Current research involves the use of chondrocytes or progenitor stem cells, associated with "smart" biomaterials and growth factors. Other influential factors, such as cell sources, oxygen pressure and mechanical strain are considered, as are recent developments in gene therapy to control the chondrocyte differentiation/dedifferentiation process. GENERAL SIGNIFICANCE This review provides new information on the mechanisms regulating the state of differentiation of chondrocytes and the chondrogenesis of mesenchymal stem cells that will lead to the development of new restorative cell therapy approaches in humans. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
|
81
|
Guerrero F, Herencia C, Almadén Y, Martínez-Moreno JM, Montes de Oca A, Rodriguez-Ortiz ME, Diaz-Tocados JM, Canalejo A, Florio M, López I, Richards WG, Rodriguez M, Aguilera-Tejero E, Muñoz-Castañeda JR. TGF-β prevents phosphate-induced osteogenesis through inhibition of BMP and Wnt/β-catenin pathways. PLoS One 2014; 9:e89179. [PMID: 24586576 PMCID: PMC3937350 DOI: 10.1371/journal.pone.0089179] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/16/2014] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) is a key cytokine during differentiation of mesenchymal stem cells (MSC) into vascular smooth muscle cells (VSMC). High phosphate induces a phenotypic transformation of vascular smooth muscle cells (VSMC) into osteogenic-like cells. This study was aimed to evaluate signaling pathways involved during VSMC differentiation of MSC in presence or not of high phosphate. RESULTS Our results showed that TGF-β induced nuclear translocation of Smad3 as well as the expression of vascular smooth muscle markers, such as smooth muscle alpha actin, SM22α, myocardin, and smooth muscle-myosin heavy chain. The addition of high phosphate to MSC promoted nuclear translocation of Smad1/5/8 and the activation of canonical Wnt/β-catenin in addition to an increase in BMP-2 expression, calcium deposition and alkaline phosphatase activity. The administration of TGF-β to MSC treated with high phosphate abolished all these effects by inhibiting canonical Wnt, BMP and TGF-β pathways. A similar outcome was observed in high phosphate-treated cells after the inhibition of canonical Wnt signaling with Dkk-1. Conversely, addition of both Wnt/β-catenin activators CHIR98014 and lithium chloride enhanced the effect of high phosphate on BMP-2, calcium deposition and alkaline phosphatase activity. CONCLUSIONS Full VSMC differentiation induced by TGF-β may not be achieved when extracellular phosphate levels are high. Moreover, TGF-β prevents high phosphate-induced osteogenesis by decreasing the nuclear translocation of Smad 1/5/8 and avoiding the activation of Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Fátima Guerrero
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | - Carmen Herencia
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | - Yolanda Almadén
- Lipid and Atherosclerosis Unit. IMIBIC/Reina Sofía University Hospital/University of Córdoba, and CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain
| | - Julio M. Martínez-Moreno
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | - Addy Montes de Oca
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | | | - Juan M. Diaz-Tocados
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | - Antonio Canalejo
- Departamento de Biología Ambiental y Salud Pública, Universidad de Huelva, Huelva, Spain
| | - Mónica Florio
- Metabolic Disorders Department, Amgen, Inc. One Amgen Center Drive, Thousand Oaks, California, United States of America
| | - Ignacio López
- Department of Medicina y Cirugía Animal, Universidad de Cordoba, Cordoba, Spain
| | - William G. Richards
- Metabolic Disorders Department, Amgen, Inc. One Amgen Center Drive, Thousand Oaks, California, United States of America
| | - Mariano Rodriguez
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
| | | | - Juan R. Muñoz-Castañeda
- Instituto Maimónides para la Investigación Biomédica (IMIBIC). Hospital Reina Sofía. Universidad de Córdoba, Córdoba, Spain
- * E-mail:
| |
Collapse
|
82
|
Liu S, Song W, Boulanger JH, Tang W, Sabbagh Y, Kelley B, Gotschall R, Ryan S, Phillips L, Malley K, Cao X, Xia TH, Zhen G, Cao X, Ling H, Dechow PC, Bellido TM, Ledbetter SR, Schiavi SC. Role of TGF-β in a mouse model of high turnover renal osteodystrophy. J Bone Miner Res 2014; 29:1141-57. [PMID: 24166835 PMCID: PMC4076799 DOI: 10.1002/jbmr.2120] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 09/24/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Altered bone turnover is a key pathologic feature of chronic kidney disease-mineral and bone disorder (CKD-MBD). Expression of TGF-β1, a known regulator of bone turnover, is increased in bone biopsies from individuals with CKD. Similarly, TGF-β1 mRNA and downstream signaling is increased in bones from jck mice, a model of high-turnover renal osteodystrophy. A neutralizing anti-TGF-β antibody (1D11) was used to explore TGF-β's role in renal osteodystrophy. 1D11 administration to jck significantly attenuated elevated serum osteocalcin and type I collagen C-telopeptides. Histomorphometric analysis indicated that 1D11 administration increased bone volume and suppressed the elevated bone turnover in a dose-dependent manner. These effects were associated with reductions in osteoblast and osteoclast surface areas. Micro-computed tomography (µCT) confirmed the observed increase in trabecular bone volume and demonstrated improvements in trabecular architecture and increased cortical thickness. 1D11 administration was associated with significant reductions in expression of osteoblast marker genes (Runx2, alkaline phosphatase, osteocalcin) and the osteoclast marker gene, Trap5. Importantly, in this model, 1D11 did not improve kidney function or reduce serum parathyroid hormone (PTH) levels, indicating that 1D11 effects on bone are independent of changes in renal or parathyroid function. 1D11 also significantly attenuated high-turnover bone disease in the adenine-induced uremic rat model. Antibody administration was associated with a reduction in pSMAD2/SMAD2 in bone but not bone marrow as assessed by quantitative immunoblot analysis. Immunostaining revealed pSMAD staining in osteoblasts and osteocytes but not osteoclasts, suggesting 1D11 effects on osteoclasts may be indirect. Immunoblot and whole genome mRNA expression analysis confirmed our previous observation that repression of Wnt/β-catenin expression in bone is correlated with increased osteoclast activity in jck mice and bone biopsies from CKD patients. Furthermore, our data suggest that elevated TGF-β may contribute to the pathogenesis of high-turnover disease partially through inhibition of β-catenin signaling.
Collapse
Affiliation(s)
- Shiguang Liu
- Genzyme, Sanofi-Genzyme R&D Center, Framingham, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Tu B, Peng ZX, Fan QM, Du L, Yan W, Tang TT. Osteosarcoma cells promote the production of pro-tumor cytokines in mesenchymal stem cells by inhibiting their osteogenic differentiation through the TGF-β/Smad2/3 pathway. Exp Cell Res 2013; 320:164-73. [PMID: 24183998 DOI: 10.1016/j.yexcr.2013.10.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/15/2013] [Accepted: 10/23/2013] [Indexed: 01/23/2023]
Abstract
Mesenchymal stem cells (MSCs) are among the most important components of the osteosarcoma microenvironment and are reported to promote tumor progression. However, the means by which osteosarcoma cells modulate MSC behavior remains unclear. The aim of this study was to determine the effects of osteosarcoma cells on both the production of pro-tumor cytokines by mesenchymal stem cells (MSCs) and the osteogenic differentiation of MSCs. High level of transforming growth factor-β (TGF-β) was detected in three osteosarcoma cell lines. Conditioned media (CM) from the osteosarcoma cell lines Saos-2 and U2-OS were used to stimulate the cultured MSCs. We found that osteosarcoma cells promoted the production of IL-6 and VEGF in MSCs by inhibiting their osteogenic differentiation. Furthermore, TGF-β in tumor CM was proved to be an important factor. The TGF-β neutralizing antibody antagonized the effects induced by osteosarcoma CM. The inhibition of Smad2/3 by siRNA significantly decreased the production of IL-6 and VEGF in MSCs and induced their osteogenic differentiation. We also found that Smad2/3 enhanced the expression of β-catenin in MSCs by decreasing the level of Dickkopf-1 (DKK1). Although the inhibition of β-catenin did not affect the production of IL-6 or VEGF, or the gene expression of the early osteogenic markers Runx2 and ALP, it did enhance the gene expression of osteocalcin. Taken together, our data indicate that osteosarcoma cells secrete TGF-β to maintain the stemness of MSCs and promote the production of pro-tumor cytokines by these cells.
Collapse
Affiliation(s)
- Bing Tu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road 639, Shanghai 200011, China
| | | | | | | | | | | |
Collapse
|
84
|
Fakhry M, Hamade E, Badran B, Buchet R, Magne D. Molecular mechanisms of mesenchymal stem cell differentiation towards osteoblasts. World J Stem Cells 2013; 5:136-148. [PMID: 24179602 PMCID: PMC3812518 DOI: 10.4252/wjsc.v5.i4.136] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/01/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic tissue that is constantly renewed by the coordinated action of two cell types, i.e., the bone-resorbing osteoclasts and the bone-forming osteoblasts. However, in some circumstances, bone regeneration exceeds bone self repair capacities. This is notably often the case after bone fractures, osteolytic bone tumor surgery, or osteonecrosis. In this regard, bone tissue engineering with autologous or allogenic mesenchymal stem cells (MSCs) is been widely developed. MSCs can be isolated from bone marrow or other tissues such as adipose tissue or umbilical cord, and can be implanted in bone defects with or without prior amplification and stimulation. However, the outcome of most pre-clinical studies remains relatively disappointing. A better understanding of the successive steps and molecular mechanisms involved in MSC-osteoblastic differentiation appears to be crucial to optimize MSC-bone therapy. In this review, we first present the important growth factors that stimulate osteoblastogenesis. Then we review the main transcription factors that modulate osteoblast differentiation, and the microRNAs (miRs) that inhibit their expression. Finally, we also discuss articles dealing with the use of these factors and miRs in the development of new bone MSC therapy strategies. We particularly focus on the studies using human MSCs, since significant differences exist between osteoblast differentiation mechanisms in humans and mice for instance.
Collapse
|
85
|
Abstract
The bone marrow milieu comprising both hematopoietic and nonhematopoietic lineages has a unique structural organization. Bone undergoes continuous remodeling throughout life. This dynamic process involves a balance between bone-forming osteoblasts (OBs) derived from multipotent mesenchymal stem cells (MSCs) and bone-resorbing osteoclasts (OCs) derived from hematopoietic stem cells (HSCs). Src homology 2-domain-containing inositol 5'-phosphatase 1 (SHIP1) regulates cellular processes such as proliferation, differentiation, and survival via the PI3K/Akt signaling pathway initiated at the plasma membrane. SHIP1-deficient mice also exhibit profound osteoporosis that has been proposed to result from hyperresorptive activity by OCs. We have previously observed that SHIP1 is expressed in primary OBs, which display defective development in SHIP1-deficient mice. These findings led us to question whether SHIP1 plays a functional role in osteolineage development from MSC in vivo, which contributes to the osteoporotic phenotype in germline SHIP1 knockout mice. In this short review, we discuss our current understanding of inositol phospholipid signaling downstream of SHIP1 in bone biology.
Collapse
Affiliation(s)
- Sonia Iyer
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
86
|
Li H, Usas A, Poddar M, Chen CW, Thompson S, Ahani B, Cummins J, Lavasani M, Huard J. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8:e64923. [PMID: 23762264 PMCID: PMC3676442 DOI: 10.1371/journal.pone.0064923] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/20/2013] [Indexed: 01/01/2023] Open
Abstract
Human muscle-derived progenitor cells (hMDPCs) offer great promise for muscle cell-based regenerative medicine; however, prolonged ex-vivo expansion using animal sera is necessary to acquire sufficient cells for transplantation. Due to the risks associated with the use of animal sera, the development of a strategy for the ex vivo expansion of hMDPCs is required. The purpose of this study was to investigate the efficacy of using platelet-rich plasma (PRP) for the ex-vivo expansion of hMDPCs. Pre-plated MDPCs, myoendothelial cells, and pericytes are three populations of hMDPCs that we isolated by the modified pre-plate technique and Fluorescence Activated Cell Sorting (FACS), respectively. Pooled allogeneic human PRP was obtained from a local blood bank, and the effect that thrombin-activated PRP-releasate supplemented media had on the ex-vivo expansion of the hMDPCs was tested against FBS supplemented media, both in vitro and in vivo. PRP significantly enhanced short and long-term cell proliferation, with or without FBS supplementation. Antibody-neutralization of PDGF significantly blocked the mitogenic/proliferative effects that PRP had on the hMDPCs. A more stable and sustained expression of markers associated with stemness, and a decreased expression of lineage specific markers was observed in the PRP-expanded cells when compared with the FBS-expanded cells. The in vitro osteogenic, chondrogenic, and myogenic differentiation capacities of the hMDPCs were not altered when expanded in media supplemented with PRP. All populations of hMDPCs that were expanded in PRP supplemented media retained their ability to regenerate myofibers in vivo. Our data demonstrated that PRP promoted the proliferation and maintained the multi-differentiation capacities of the hMDPCs during ex-vivo expansion by maintaining the cells in an undifferentiated state. Moreover, PDGF appears to be a key contributing factor to the beneficial effect that PRP has on the proliferation of hMDPCs.
Collapse
Affiliation(s)
- Hongshuai Li
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Arvydas Usas
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Minakshi Poddar
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chien-Wen Chen
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Seth Thompson
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bahar Ahani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - James Cummins
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mitra Lavasani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Johnny Huard
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
87
|
Kazemnejad S, Khanmohammadi M, Zarnani A, Nikokar I, Saghari S. Role of Wnt signaling on proliferation of menstrual blood derived stem cells. J Stem Cells Regen Med 2013. [PMID: 24693204 PMCID: PMC3908310 DOI: 10.46582/jsrm.0901004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIM Menstrual blood derived stem cells (MenSCs) are unique stem cells that have been isolated and identified recently. The special traits of MenSCs can be related to the cell signaling pathways. In this study, in order to find out the role of Wnt signaling on MenSCs proliferation, we evaluated ß-catenin expression as a key participant in Wnt signaling pathway in response to Lithium chloride (LiCl). METHODS MenSCs were isolated from healthy women by combining gradient density centrifugation with plastic adherence. After characterization of the isolated cells, cell proliferation of MenSCs in presence of 10-15 mM LiCl was evaluated by MTT assay. ß-catenin expression of the treated cells was examined using immunofluorescence technique. RESULTS Flow cytometric analysis revealed that both mesenchymal and embryonic stem cell markers are expressed on menstrual blood stem cells. MTT value decreased depending on the LiCl concentration. The proliferation of MenSCs cultivated in culture media containing 15mM LiCl was approximately two fold less than those grown without LiCl (p<0.01). Moreover, nuclear accumulation of ß-catenin protein in cells treated by LiCl was greater than cells without LiCl. CONCLUSION The MenSCs are stem cell populations with high proliferation ability and unique immunophenotyping properties. Our results demonstrated that Wnt signaling pathway regulates MenSCs proliferation via trans-localization of activated-ß-catenin protein.
Collapse
Affiliation(s)
- S Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute , ACECR, Tehran, Iran . ; Paramedical Faculty of Guilan University of Medical Sciences , Langroud, Guilan, Iran
| | - M Khanmohammadi
- Reproductive Biotechnology Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Ah Zarnani
- Nanobiotechnology Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - I Nikokar
- Paramedical Faculty of Guilan University of Medical Sciences , Langroud, Guilan, Iran
| | - S Saghari
- Paramedical Faculty of Guilan University of Medical Sciences , Langroud, Guilan, Iran
| |
Collapse
|
88
|
Zhou S, Mizuno S, Glowacki J. Wnt pathway regulation by demineralized bone is approximated by both BMP-2 and TGF-β1 signaling. J Orthop Res 2013; 31:554-60. [PMID: 23239467 DOI: 10.1002/jor.22244] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 09/14/2012] [Indexed: 02/04/2023]
Abstract
Allogeneic demineralized bone is used extensively as a clinical graft material because it has osteo/chondroinductive and osteoconductive properties. Demineralized bone powder (DBP) induces chondrogenic differentiation of human dermal fibroblasts (hDFs) in three-dimensional collagen cultures, but the initiating mechanisms have not been fully characterized nor has it been shown that bone morphogenetic proteins (BMPs) recapitulate DBP's effects on target cells. Among the many signaling pathways regulated in hDFs by DBP prior to in vitro chondrogenesis, there are changes in Wnts and their receptors that may contribute to DBP actions. This study tests the hypothesis that DBP modulation of Wnt signaling entails both BMP and TGF-β pathways. We compared the effects of DBP, TGF-β1, or BMP-2 on Wnt signaling components in hDFs by Wnt signaling macroarray, RT-PCR, in situ hybridization, and Western immunoblot analyses. Many effects of DBP on Wnt signaling components were not shared by BMP-2, and likewise DBP effects on Wnt genes and β-catenin only partially required the TGF-β pathway, as shown by selective inhibition of TGF-β/activin receptor-like kinase. The analyses revealed that 64% (16/25) of the Wnt signaling components regulated by DBP were regulated similarly by the sum of effects by BMP-2 and by TGF-β1. In conclusion, signaling mechanisms of inductive DBP in human dermal fibroblasts involve the modulation of multiple Wnt signals through both BMP and TGF-β pathways.
Collapse
Affiliation(s)
- Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
89
|
Kasagi S, Chen W. TGF-beta1 on osteoimmunology and the bone component cells. Cell Biosci 2013; 3:4. [PMID: 23321200 PMCID: PMC3565958 DOI: 10.1186/2045-3701-3-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/26/2012] [Indexed: 01/07/2023] Open
Abstract
TGF-β1 is an immunoregulatory cytokine that regulates immune cell proliferation, survival, differentiation, and migration. Compelling evidence has demonstrated a strong association between the immune and skeletal systems (so called Osteoimmunology), such as the critical role of TGF-β1 in the development and maintenance of the skeletal tissue. This review provides an overview of the mechanisms in which TGF-β1 interacts with bone component cells, such as osteoblasts, osteoclasts, chondrocytes, mesenchymal stem cells, and hematopoietic stem cells, in concert with other cytokines and hormones.
Collapse
Affiliation(s)
- Shimpei Kasagi
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, USA.
| | | |
Collapse
|
90
|
Maeda H, Wada N, Tomokiyo A, Monnouchi S, Akamine A. Prospective potency of TGF-β1 on maintenance and regeneration of periodontal tissue. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:283-367. [PMID: 23809439 DOI: 10.1016/b978-0-12-407696-9.00006-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Periodontal ligament (PDL) tissue, central in the periodontium, plays crucial roles in sustaining tooth in the bone socket. Irreparable damages of this tissue provoke tooth loss, causing a decreased quality of life. The question arises as to how PDL tissue is maintained or how the lost PDL tissue can be regenerated. Stem cells included in PDL tissue (PDLSCs) are widely accepted to have the potential to maintain or regenerate the periodontium, but PDLSCs are very few in number. In recent studies, undifferentiated clonal human PDL cell lines were developed to elucidate the applicable potentials of PDLSCs for the periodontal regenerative medicine based on cell-based tissue engineering. In addition, it has been suggested that transforming growth factor-beta 1 is an eligible factor for the maintenance and regeneration of PDL tissue.
Collapse
Affiliation(s)
- Hidefumi Maeda
- Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
91
|
Benisch P, Schilling T, Klein-Hitpass L, Frey SP, Seefried L, Raaijmakers N, Krug M, Regensburger M, Zeck S, Schinke T, Amling M, Ebert R, Jakob F. The transcriptional profile of mesenchymal stem cell populations in primary osteoporosis is distinct and shows overexpression of osteogenic inhibitors. PLoS One 2012; 7:e45142. [PMID: 23028809 PMCID: PMC3454401 DOI: 10.1371/journal.pone.0045142] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 08/13/2012] [Indexed: 12/11/2022] Open
Abstract
Primary osteoporosis is an age-related disease characterized by an imbalance in bone homeostasis. While the resorptive aspect of the disease has been studied intensely, less is known about the anabolic part of the syndrome or presumptive deficiencies in bone regeneration. Multipotent mesenchymal stem cells (MSC) are the primary source of osteogenic regeneration. In the present study we aimed to unravel whether MSC biology is directly involved in the pathophysiology of the disease and therefore performed microarray analyses of hMSC of elderly patients (79–94 years old) suffering from osteoporosis (hMSC-OP). In comparison to age-matched controls we detected profound changes in the transcriptome in hMSC-OP, e.g. enhanced mRNA expression of known osteoporosis-associated genes (LRP5, RUNX2, COL1A1) and of genes involved in osteoclastogenesis (CSF1, PTH1R), but most notably of genes coding for inhibitors of WNT and BMP signaling, such as Sclerostin and MAB21L2. These candidate genes indicate intrinsic deficiencies in self-renewal and differentiation potential in osteoporotic stem cells. We also compared both hMSC-OP and non-osteoporotic hMSC-old of elderly donors to hMSC of ∼30 years younger donors and found that the transcriptional changes acquired between the sixth and the ninth decade of life differed widely between osteoporotic and non-osteoporotic stem cells. In addition, we compared the osteoporotic transcriptome to long term-cultivated, senescent hMSC and detected some signs for pre-senescence in hMSC-OP. Our results suggest that in primary osteoporosis the transcriptomes of hMSC populations show distinct signatures and little overlap with non-osteoporotic aging, although we detected some hints for senescence-associated changes. While there are remarkable inter-individual variations as expected for polygenetic diseases, we could identify many susceptibility genes for osteoporosis known from genetic studies. We also found new candidates, e.g. MAB21L2, a novel repressor of BMP-induced transcription. Such transcriptional changes may reflect epigenetic changes, which are part of a specific osteoporosis-associated aging process.
Collapse
Affiliation(s)
- Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Tatjana Schilling
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Ludger Klein-Hitpass
- Institute of Cell Biology (Tumor Research), University Hospital Essen, Essen, Germany
| | - Sönke P. Frey
- Department of Trauma, Hand-, Plastic- and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Nadja Raaijmakers
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Melanie Krug
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Regensburger
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
92
|
Saeed H, Taipaleenmäki H, Aldahmash AM, Abdallah BM, Kassem M. Mouse embryonic fibroblasts (MEF) exhibit a similar but not identical phenotype to bone marrow stromal stem cells (BMSC). Stem Cell Rev Rep 2012; 8:318-28. [PMID: 21927803 DOI: 10.1007/s12015-011-9315-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mouse embryonic fibroblasts have been utilized as a surrogate stem cell model for the postnatal bone marrow-derived stromal stem cells (BMSC) to study mesoderm-type cell differentiation e.g. osteoblasts, adipocytes and chondrocytes. However, no formal characterization of MEF phenotype has been reported. Utilizing standard in vitro and in vivo assays we performed a side-by-side comparison of MEF and BMSC to determine their ability to differentiate into mesoderm-type cells. BMSC were isolated from 8-10 weeks old mouse bone marrow by plastic adherence. MEF were established by trypsin/EDTA digestion from E13.5 embryos after removing heads and viscera, followed by plastic adherence. Compared to BMSC, MEF exhibited telomerase activity and improved cell proliferation as assessed by q-PCR based TRAP assay and cell number quantification, respectively. FACS analysis revealed that MEF exhibited surface markers characteristic of the BMSC: Sca-1(+), CD73(+), CD105(+), CD29(+), CD44(+), CD106(+), CD11b(-), and CD45(-). In contrast to BMSC, ex vivo osteoblast (OB) differentiation of MEF exhibited a less mature osteoblastic phenotype (less alkaline phosphatase, collagen type I and osteocalcin) as assessed by real-time PCR analysis. Compared to BMSC, MEF exhibited a more enhanced differentiation into adipocyte and chondrocyte lineages. Interestingly, both MEF and BMSC formed the same amount of heterotopic bone and bone marrow elements upon in vivo subcutaneous implantation with hydroxyapatite/tricalcium phosphate, in immune deficient mice. In conclusion, MEF contain a population of stem cells that behave in ex vivo and in vivo assays, similar but not identical, to BMSC. Due to their enhanced cell growth, they may represent a good alternative for BMSC in studying molecular mechanisms of stem cell commitment and differentiation to osteoblasts, adipocytes and chondrocytes.
Collapse
Affiliation(s)
- Hamid Saeed
- Endocrine Research Laboratory, KMEB, Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | | | | | | | | |
Collapse
|
93
|
Narcisi R, Quarto R, Ulivi V, Muraglia A, Molfetta L, Giannoni P. TGF β-1 administration during ex vivo expansion of human articular chondrocytes in a serum-free medium redirects the cell phenotype toward hypertrophy. J Cell Physiol 2012; 227:3282-90. [PMID: 22105490 DOI: 10.1002/jcp.24024] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell-based cartilage resurfacing requires ex vivo expansion of autologous articular chondrocytes. Defined culture conditions minimize expansion-dependent phenotypic alterations but maintenance of the cells' differentiation potential must be carefully assessed. Transforming growth factor β-1 (TGF β-1) positively regulates the expression of several cartilage proteins, but its therapeutic application in damaged cartilage is controversial. Thus we evaluated the phenotypic outcomes of cultured human articular chondrocytes exposed to TGF β-1 during monolayer expansion in a serum-free medium. After five doublings cells were transferred to micromass cultures to assess their chondrogenic differentiation, or replated in osteogenic medium. Immunocytostainings of micromasses of TGF-expanded cells showed loss of aggrecan and type II collagen. Positivity was evidenced for RAGE, IHH, type X collagen and for apoptotic cells, paralleling a reduction of BCL-2 levels, suggesting hypertrophic differentiation. TGF β-1-exposed cells also evidenced increased mRNA levels for bone sialoprotein, osteopontin, matrix metalloproteinase-13, TIMP-3, VEGF and SMAD7, enhanced alkaline phosphatase activity and pyrophosphate availability. Conversely, SMAD3 mRNA and protein contents were reduced. After osteogenic induction, only TGF-expanded cells strongly mineralized and impaired p38 kinase activity, a contributor of chondrocytes' differentiation. To evaluate possible endochondral ossification progression, we seeded the chondrocytes on hydroxyapatite scaffolds, subsequently implanted in an in vivo ectopic setting, but cells failed to reach overt ossification; nonetheless, constructs seeded with TGF-exposed cells displayed blood vessels of the host vascular supply with enlarged diameters, suggestive of vascular remodeling, as in bone growth. Thus TGF-exposure during articular chondrocytes expansion induces a phenotype switch to hypertrophy, an undesirable effect for cells possibly intended for tissue-engineered cartilage repair.
Collapse
Affiliation(s)
- R Narcisi
- Stem Cell Laboratory, Advanced Biotechnology Center, Genova, Italy
| | | | | | | | | | | |
Collapse
|
94
|
Kim YM, Kim J, Heo SC, Shin SH, Do EK, Suh DS, Kim KH, Yoon MS, Lee TG, Kim JH. Proteomic identification of ADAM12 as a regulator for TGF-β1-induced differentiation of human mesenchymal stem cells to smooth muscle cells. PLoS One 2012; 7:e40820. [PMID: 22808268 PMCID: PMC3396647 DOI: 10.1371/journal.pone.0040820] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/13/2012] [Indexed: 12/02/2022] Open
Abstract
Background Transforming growth factor-β1 (TGF-β1) induces the differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) into smooth muscle cells. Lipid rafts are cholesterol-rich microdomains in cell membranes that reportedly play a key role in receptor-mediated signal transduction and cellular responses. In order to clarify whether lipid rafts are involved in TGF-β1-induced differentiation of hASCs into smooth muscle cells, we analyzed the lipid raft proteome of hASCs. Methods and Results Pretreatment of hASCs with the lipid raft disruptor methyl-β-cyclodextrin abrogated TGF-β1-induced expression of α-smooth muscle actin, a smooth muscle cell marker, suggesting a pivotal role of lipid rafts in TGF-β1-induced differentiation of hASCs to smooth muscle cells. Sucrose density gradient centrifugation along with a shotgun proteomic strategy using liquid chromatography-tandem mass spectrometry identified 1002 individual proteins as the lipid raft proteome, and 242 of these were induced by TGF-β1 treatment. ADAM12, a disintegrin and metalloproteases family member, was identified as the most highly up-regulated protein in response to TGF-β1 treatment. TGF-β1 treatment of hASCs stimulated the production of both ADAM12 protein and mRNA. Silencing of endogenous ADAM12 expression using lentiviral small hairpin RNA or small interfering RNA abrogated the TGF-β1-induced differentiation of hASCs into smooth muscle cells. Conclusions These results suggest a pivotal role for lipid raft-associated ADAM12 in the TGF-β1-induced differentiation of hASCs into smooth muscle cells.
Collapse
Affiliation(s)
- Young Mi Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jaeyoon Kim
- NovaCell Technology Inc., Pohang, Republic of Korea
| | - Soon Chul Heo
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sang Hun Shin
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Eun Kyoung Do
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Dong-Soo Suh
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ki-Hyung Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Man-Soo Yoon
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | | | - Jae Ho Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- * E-mail:
| |
Collapse
|
95
|
Bailey Dubose K, Zayzafoon M, Murphy-Ullrich JE. Thrombospondin-1 inhibits osteogenic differentiation of human mesenchymal stem cells through latent TGF-β activation. Biochem Biophys Res Commun 2012; 422:488-93. [PMID: 22583901 DOI: 10.1016/j.bbrc.2012.05.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 05/04/2012] [Indexed: 12/31/2022]
Abstract
Transforming growth factor-β (TGF-β) is a critical regulator of bone development and remodeling. TGF-β must be activated from its latent form in order to signal. Thrombospondin-1 (TSP1) is a major regulator of latent TGF-β activation and TSP1 control of TGF-β activation is critical for regulation of TGF-β activity in multiple diseases. Bone marrow-derived mesenchymal stem cells (MSCs) have osteogenic potential and they participate in bone remodeling in injury and in response to tumor metastasis. Since both TSP1 and TGF-β inhibit osteoblast differentiation, we asked whether TSP1 blocks osteoblast differentiation of MSCs through its ability to stimulate TGF-β activation. TSP1 added to human bone marrow-derived MSCs under growth conditions increases active TGF-β. Cultured MSCs express TSP1 and both TSP1 expression and TGF-β activity decrease during osteoblast differentiation. TSP1 and active TGF-β block osteoblast differentiation of MSCs grown in osteogenic media as measured by decreased Runx2 and alkaline phosphatase expression. The inhibitory effect of TSP1 on osteoblast differentiation is due to its ability to activate latent TGF-β, since a peptide which blocks TSP1 TGF-β activation reduced TGF-β activity and restored osteoblast differentiation as measured by increased Runx2 and alkaline phosphatase expression. Anti-TGF-β neutralizing antibody also increased alkaline phosphatase expression in the presence of TSP1. These studies show that TSP1 regulated TGF-β activity is a critical determinant of osteoblast differentiation.
Collapse
Affiliation(s)
- Kimberly Bailey Dubose
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA.
| | | | | |
Collapse
|
96
|
Bakhshandeh B, Soleimani M, Hafizi M, Paylakhi SH, Ghaemi N. MicroRNA signature associated with osteogenic lineage commitment. Mol Biol Rep 2012; 39:7569-81. [PMID: 22350160 DOI: 10.1007/s11033-012-1591-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/31/2012] [Indexed: 01/22/2023]
Abstract
Cell-based approaches offer a potential therapeutic strategy for appropriate bone manufacturing. Capable of differentiating into multiple cell types especially osteoblasts spontaneously, unrestricted somatic stem cell (USSC) seems to be a suitable candidate. Recent studies have shown the involvement of microRNAs in several biological processes. miRNA microarray profiling was applied in order to identify the osteo-specific miRNA signature. Prior to this analysis, osteogenic commitment of osteoblasts was evaluated by measuring ALPase activity, biomineralization, specific staining and evaluation of some main osteogenic marker genes. To support our findings, various in silico explorations (for both putative targets and signaling pathways) and empirical analyses (miRNA transfections followed by qPCR of osteogenic indicators and ALPase activity measurement) were carried out. The function of GSK-3b inhibitor was also studied to investigate the role of WNT in osteogenesis. Transient modulation of multiple osteo-miRs (such as mir-199b, 1274a, 30b) with common targets (such as BMPR, TCFs, SMADs) as mediators of osteogenic pathways including cell-cell interactions, WNT and TGF-beta pathways, suggests a mechanism for rapid induction of the osteogenesis as an anti-miRNA therapy. The results of this research have identified the miRNA signature which regulates the osteogenesis mechanism in USSC. To conclude, our study reveals more details about the allocation of USSCs into osteogenic lineage through modulatory effect of miRNAs on targets and pathways required for creating a tissue-specific phenotype and may aid in future clinical interventions.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | | | | | | | | |
Collapse
|
97
|
Chen G, Deng C, Li YP. TGF-β and BMP signaling in osteoblast differentiation and bone formation. Int J Biol Sci 2012; 8:272-88. [PMID: 22298955 PMCID: PMC3269610 DOI: 10.7150/ijbs.2929] [Citation(s) in RCA: 1319] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 12/29/2011] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta (TGF-β)/bone morphogenic protein (BMP) signaling is involved in a vast majority of cellular processes and is fundamentally important throughout life. TGF-β/BMPs have widely recognized roles in bone formation during mammalian development and exhibit versatile regulatory functions in the body. Signaling transduction by TGF-β/BMPs is specifically through both canonical Smad-dependent pathways (TGF-β/BMP ligands, receptors and Smads) and non-canonical Smad-independent signaling pathway (e.g. p38 mitogen-activated protein kinase pathway, MAPK). Following TGF-β/BMP induction, both the Smad and p38 MAPK pathways converge at the Runx2 gene to control mesenchymal precursor cell differentiation. The coordinated activity of Runx2 and TGF-β/BMP-activated Smads is critical for formation of the skeleton. Recent advances in molecular and genetic studies using gene targeting in mice enable a better understanding of TGF-β/BMP signaling in bone and in the signaling networks underlying osteoblast differentiation and bone formation. This review summarizes the recent advances in our understanding of TGF-β/BMP signaling in bone from studies of genetic mouse models and human diseases caused by the disruption of TGF-β/BMP signaling. This review also highlights the different modes of cross-talk between TGF-β/BMP signaling and the signaling pathways of MAPK, Wnt, Hedgehog, Notch, and FGF in osteoblast differentiation and bone formation.
Collapse
Affiliation(s)
- Guiqian Chen
- Institute of Genetics, Life Science College, Zhejiang University, 388 Yuhang Road, Hangzhou 310058, China
| | | | | |
Collapse
|
98
|
Microarray-based bioinformatics analysis of osteoblasts on TiO2 nanotube layers. Colloids Surf B Biointerfaces 2011; 93:135-42. [PMID: 22261177 DOI: 10.1016/j.colsurfb.2011.12.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 12/22/2011] [Accepted: 12/22/2011] [Indexed: 12/17/2022]
Abstract
The TiO(2) nanotube layers fabricated by electrochemical anodization have received considerable attention in dentistry and orthopedic medicine due to their increased osseointegration compared with the unanodized titanium. The molecular mechanisms underlying the interactions between nanotubes and osteoblasts is unknown. To examine this, the mRNA expression profile of MG-63 osteoblast-like cells cultured on the TiO(2) nanotubes was explored by DNA microarray. The differentially expressed genes were identified by bioinformatics analysis. Gene ontology (GO) and Go-map network analysis indicated that the TiO(2) nanotubes enhanced osteoblast proliferation and differentiation and decreased osteoblast adhesion and immunization. The expressions of genes were mainly increased in pathways influencing cell proliferation and differentiation (Cell cycle, Terpenoid backbone biosynthesis, and TGF-beta signaling) and were decreased in pathways controlling cell immunization (Cell adhesion molecules (CAMs), Allograft rejection, and Graft-versus-host disease). Signal network analysis generated from differentially expressed genes suggested that CTNNB1 (beta-catenin) was the central gene for increasing osteoblast proliferation and differentiation, and IKBKG (inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma) was the central gene for repressing osteoblast immunization on nanotube layers. These two genes were further confirmed by quantitative PCR. The identified signal pathways and central genes in the study are well correlated with osteoblast phenotype. Furthermore, microarray-based bioinformatics analysis is a powerful tool in efficiently understanding molecular mechanisms underlying the interactions between osteoblasts and the nanotube layers.
Collapse
|
99
|
Geng S, Zhou S, Glowacki J. Age-related decline in osteoblastogenesis and 1α-hydroxylase/CYP27B1 in human mesenchymal stem cells: stimulation by parathyroid hormone. Aging Cell 2011; 10:962-71. [PMID: 21824271 DOI: 10.1111/j.1474-9726.2011.00735.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With aging, there is a decline in bone mass and in osteoblast differentiation of human mesenchymal stem cells (hMSCs) in vitro. Osteoblastogenesis can be stimulated with 1,25-dihydroxyvitamin D(3) [1,25(OH)(2) D(3) ] and, in some hMSCs, by the precursor 25-hydroxyvitamin D(3) (25OHD(3) ). CYP27B1/1α-hydroxylase activates 25OHD(3) and, to a variable degree, hMSCs express CYP27B1. In this study, we tested the hypotheses (i) that age affects responsiveness to 25OHD(3) and expression/activity of CYP27B1 in hMSCs and (ii) that parathyroid hormone (PTH) upregulates CYP27B1 in hMSCs, as it does in renal cells. There were age-related declines in osteoblastogenesis (n=8, P=0.0286) and in CYP27B1 gene expression (n=27, r= -0.498; P=0.008) in hMSCs. Unlike hMSCs from young subjects (≤50 years), hMSCs from older subjects (≥55 years) were resistant to 25OHD(3) stimulation of osteoblastogenesis. PTH1-34 (100 nm) provided hMSCs with responsiveness to 25OHD(3) (P=0.0313, Wilcoxon matched pairs test) and with two episodes of increased 1,25(OH)(2) D(3) synthesis, of cAMP response element binding protein (CREB) activation, and of CYP27B1 upregulation. Both increases in CYP27B1 expression by PTH were obliterated by CREB-siRNA or KG-501 (which specifically inhibits the downstream binding of activated CREB). Only the second period of CREB signaling was diminished by AG1024, an inhibitor of insulin-like growth factor-I receptor kinase. Thus, PTH stimulated hMSCs from elders with responsiveness to 25OHD(3) by upregulating expression/activity of CYP27B1 and did so through CREB and IGF-I pathways.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | | | |
Collapse
|
100
|
Wnt/β-catenin pathway forms a negative feedback loop during TGF-β1 induced human normal skin fibroblast-to-myofibroblast transition. J Dermatol Sci 2011; 65:38-49. [PMID: 22041457 DOI: 10.1016/j.jdermsci.2011.09.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 07/17/2011] [Accepted: 09/30/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND Fibroblast-to-myofibroblast transition is a key event during wound healing and hypertrophic scar formation. Previous studies suggested Wnt/β-catenin signaling might be involved in the wound healing. However, its specific role in skin fibroblast-to-myofibroblast transition remains unclear. OBJECTIVE To investigate the specific role of β-catenin during the transforming growth factor-β1 induced normal skin myofibroblasts transition. METHODS By real-time quantitative polymerase chain reaction, Western-blot and immunocytochemistry, the activation of Wnt/β-catenin pathway in cultured human normal skin fibroblasts during TGF-β1 induced fibroblast-to-myofibroblast transition was investigated. The effects of β-catenin on myofibroblasts transition were also investigated when SB-216763, over-expression and siRNA of β-catenin were utilized. In addition, fibroblasts populated collagen lattices contraction assays were conducted to examine the effects of β-catenin on the contractility of the fibroblasts induced by TGF-β1. Furthermore, the effects of β-catenin on the expression of α-smooth muscle actin and collagen types I and III in hypertrophic scar derived fibroblasts were studied. RESULTS The expression of Wnts mRNA and β-catenin protein was up-regulated by TGF-β1 stimulation during the myofibroblasts transition. Both of SB-216763 and β-catenin over-expression was paralleled with decreased expression of α-smooth muscle actin, collagen types I and III, while siRNA targeting β-catenin leads to up-regulation of α-smooth muscle actin, collagen types I and III. The increased contractility and α-smooth muscle actin expression of the fibroblasts in the collagen lattices induced by TGF-β1 was inhibited by SB-216763. In addition, the expression levels of α-smooth muscle actin, collagen types I and III in hypertrophic scar derived fibroblasts were also down-regulated by SB-216763. CONCLUSION Specifically in normal skin fibroblasts, β-catenin might be involved in the myofibroblasts transition and negatively regulate the TGF-β1-induced myofibroblast transition.
Collapse
|