151
|
Constitutive β-catenin activation in osteoblasts impairs terminal osteoblast differentiation and bone quality. Exp Cell Res 2016; 350:123-131. [PMID: 27865936 DOI: 10.1016/j.yexcr.2016.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/09/2016] [Accepted: 11/15/2016] [Indexed: 12/13/2022]
Abstract
Accumulating evidence suggests that Wnt/β-catenin signaling plays a central role in controlling bone mass. We previously reported that constitutive activation of β-catenin (CA-β-catenin) in osteoblasts potentially has side effects on the bone growth and bone remodeling process, although it could increase bone mass. The present study aimed to observe the effects of osteoblastic CA-β-catenin on bone quality and to investigate possible mechanisms of these effects. It was found that CA-β-catenin mice exhibited lower mineralization levels and disorganized collagen in long bones as confirmed by von Kossa staining and sirius red staining, respectively. Also, bone strength decreased significantly in CA-β-catenin mice. Then the effect of CA-β-catenin on biological functions of osteoblasts were investigated and it was found that the expression levels of osteocalcin, a marker for the late differentiation of osteoblasts, decreased in CA-β-catenin mice, while the expression levels of osterix and alkaline phosphatase, two markers for the early differentiation of osteoblasts, increased in CA-β-catenin mice. Furthermore, higher proliferation rate were revealed in osteoblasts that were isolated from CA-β-catenin mice. The Real-time PCR and western blot examination found that the expression level of c-myc and cyclin D1, two G1 progression-related molecules, increased in osteoblasts that were isolated from the CA-β-catenin mice, and the expression levels of CDK14 and cyclin Y, two mitotic-related molecules that can accelerate cells entering into S and G2/M phases, increased in osteoblasts that were isolated from the CA-β-catenin mice. In summary, osteoblastic CA-β-catenin kept osteoblasts in high proliferative state and impaired the terminal osteoblast differentiation, and this led to changed bone structure and decreased bone strength.
Collapse
|
152
|
Kristianto J, Johnson MG, Afzal R, Blank RD. WITHDRAWN: Endothelin signaling in bone. Transl Res 2016:S1931-5244(16)30366-8. [PMID: 27893988 DOI: 10.1016/j.trsl.2016.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/27/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Jasmin Kristianto
- Endocrine and Reproductive Physiology Program, University of Wisconsin, Madison, Wis; Geriatrics Research, Education, and Clinical Center, William S. Middleton Veterans Hospital, Madison, Wis; Department of Medicine, University of Wisconsin, Madison, Wis; Division of Endocrinology, Metabolism, and Clinical Nutrition, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wis
| | - Michael G Johnson
- Endocrine and Reproductive Physiology Program, University of Wisconsin, Madison, Wis; Geriatrics Research, Education, and Clinical Center, William S. Middleton Veterans Hospital, Madison, Wis; Department of Medicine, University of Wisconsin, Madison, Wis
| | - Rafia Afzal
- Department of Anesthesiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Robert D Blank
- Endocrine and Reproductive Physiology Program, University of Wisconsin, Madison, Wis; Geriatrics Research, Education, and Clinical Center, William S. Middleton Veterans Hospital, Madison, Wis; Division of Endocrinology, Metabolism, and Clinical Nutrition, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wis; Medical Service, Clement J. Zablocki VAMC, Milwaukee, Wis
| |
Collapse
|
153
|
Aminuddin NI, Ahmad R, Akbar SA, Pingguan-Murphy B. Osteoblast and stem cell response to nanoscale topographies: a review. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2016; 17:698-714. [PMID: 27933112 PMCID: PMC5127258 DOI: 10.1080/14686996.2016.1242999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 06/06/2023]
Abstract
To understand how cells respond to the nanoscale extracellular environment in vivo, cells from various sources have been cultured on nanoscale patterns fabricated using bottom-up and top-down techniques. Human fetal osteoblasts (hFOBs) and stem cells are some of them and they are known to be overtly responsive to nanoscale topographies - allowing us to investigate the hows and whys of the response in vitro. Information gathered from these in vitro studies could be used to control the cells, i.e. make the stem cells differentiate or retain their characteristics without the use of medium supplements. In this review, hFOB and stem cell responses to nanotopographies are summarized and discussed to shed some light on the influence of patterns on the reactions. Although both types of cells are responsive to nanoscale topographies, the responses are found to be unique to topographical dimension, shape, orientation and the types of cells used. This implies that cellular responses are influenced by multitude of factors and that if done right, cheaper self-assembled nanotopographies can be tailored to control the cells. A new self-assembly, powder-based technique is also included to provide an insight into the future of nanofabrication.
Collapse
Affiliation(s)
- Nur Izzati Aminuddin
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Roslina Ahmad
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Sheikh Ali Akbar
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
154
|
Kim AY, Kim Y, Lee SH, Yoon Y, Kim WH, Kweon OK. Effect of Gelatin on Osteogenic Cell Sheet Formation Using Canine Adipose-Derived Mesenchymal Stem Cells. Cell Transplant 2016; 26:115-123. [PMID: 27725063 DOI: 10.3727/096368916x693338] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Osteogenically differentiated cell sheet techniques using mesenchymal stem cells (MSCs) are available to stimulate bone regeneration. The advantage of the cell sheet technique is delivering live cells effectively into the focal region. We developed a novel osteogenic cell sheet technique by adding gelatin to osteogenic cell medium. Gelatin-induced osteogenic cell sheets (GCSs) were compared to conventional osteogenic cell sheets (OCSs). Undifferentiated MSCs (UCs) were used as a control. The morphology of these cell sheets was evaluated microscopically and histologically. The time-dependent cell proliferation rate was estimated by DNA quantification. The expression of osteogenic gene markers and the number of calcium depositions were assessed by quantitative real-time polymerase chain reaction and Alizarin red S (ARS) staining, respectively. GCSs were thicker and stronger than OCSs. GCSs showed a significantly higher cell proliferation rate compared to OCSs (p < 0.05). GCSs exhibited significantly higher upregulation of BMP-7 mRNA compared to OCSs (p < 0.05). Both GCSs and OCSs showed negative ARS reactivity on day 10, but only GCSs showed positive ARS reactivity on day 21. With this technique, we observed active cell proliferation with abundant ECM and upregulation of osteogenic bone markers, and our results suggest that GCSs could be promising for therapeutic applications in bone regeneration.
Collapse
|
155
|
Kamiya N, Shuxian L, Yamaguchi R, Phipps M, Aruwajoye O, Adapala NS, Yuan H, Kim HKW, Feng JQ. Targeted disruption of BMP signaling through type IA receptor (BMPR1A) in osteocyte suppresses SOST and RANKL, leading to dramatic increase in bone mass, bone mineral density and mechanical strength. Bone 2016; 91:53-63. [PMID: 27402532 DOI: 10.1016/j.bone.2016.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/17/2023]
Abstract
Recent studies suggest a critical role of osteocytes in controlling skeletal development and bone remodeling although the molecular mechanism is largely unknown. This study investigated BMP signaling in osteocytes by disrupting Bmpr1a under the Dmp1-promoter. The conditional knockout (cKO) mice displayed a striking osteosclerotic phenotype with increased trabecular bone volume, thickness, number, and mineral density as assessed by X-ray and micro-CT. The bone histomorphometry, H&E, and TRAP staining revealed a dramatic increase in trabecular and cortical bone masses but a sharp reduction in osteoclast number. Moreover, there was an increase in BrdU positive osteocytes (2-5-fold) and osteoid volume (~4-fold) but a decrease in the bone formation rate (~85%) in the cKO bones, indicating a defective mineralization. The SEM analysis revealed poorly formed osteocytes: a sharp increase in cell numbers, a great reduction in cell dendrites, and a remarkable change in the cell distribution pattern. Molecular studies demonstrated a significant decrease in the Sost mRNA levels in bone (>95%), and the SOST protein levels in serum (~85%) and bone matrices. There was a significant increase in the β-catenin (>3-fold) mRNA levels as well as its target genes Tcf1 (>6-fold) and Tcf3 (~2-fold) in the cKO bones. We also showed a significant decrease in the RANKL levels of serum proteins (~65%) and bone mRNA (~57%), and a significant increase in the Opg mRNA levels (>20-fold) together with a significant reduction in the Rankl/Opg ratio (>95%), which are responsible for a sharp reduction in the cKO osteoclasts. The values of mechanical strength were higher in cKO femora (i.e. max force, displacement, and work failure). These results suggest that loss of BMP signaling specifically in osteocytes dramatically increases bone mass presumably through simultaneous inhibition of RANKL and SOST, leading to osteoclast inhibition and Wnt activation together. Finally, a working hypothesis is proposed to explain how BMPR1A controls bone remodeling by inhibiting cell proliferation and stimulating differentiation. It is reported that RANKL and SOST are abundantly expressed by osteocytes. Thus, BMP signaling through BMPR1A plays important roles in osteocytes.
Collapse
Affiliation(s)
- Nobuhiro Kamiya
- Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8883, USA; Sports Medicine, Tenri University, Tenri, Nara 632-0071, Japan.
| | - Lin Shuxian
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA; Department of Prosthodontics, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | | | - Matthew Phipps
- Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | | | - Naga Suresh Adapala
- Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8883, USA
| | - Hui Yuan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Harry K W Kim
- Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8883, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA.
| |
Collapse
|
156
|
Freise C, Kretzschmar N, Querfeld U. Wnt signaling contributes to vascular calcification by induction of matrix metalloproteinases. BMC Cardiovasc Disord 2016; 16:185. [PMID: 27716072 PMCID: PMC5045611 DOI: 10.1186/s12872-016-0362-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 09/27/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Vascular calcifications such as arteriosclerosis, which is characterized by a calcificiation of the tunica media, represent major comorbidities e.g. in patients with chronic kidney disease (CKD). An essential step during the development of arteriosclerosis is the transdifferentiation/calcification of vascular smooth muscle cells (VSMC) resembling osteogenesis. The matrix metalloproteinases (MMP)-2 and -9 were shown to promote these VSMC calcifications and their inhibition is of therapeutic value to prevent arteriosclerosis in preclinical studies. Aiming for an understanding of the underlying regulatory mechanisms of MMPs we here investigated, if the MMP-mediated VSMC calcification involves altered signaling of the Wnt pathway, which is known to impact osteogenesis. METHODS We used an experimental in vitro model of vascular calcification. Transdifferentiation/calcification of murine VSMC was induced by elevated calcium and phosphorus levels. Calcification was assessed by calcium and alkaline phosphatase measurements. Activation/activity of the gelatinases MMP-2 and MMP-9 was assessed by conversion of fluorescence-labelled substrates. Activation of the Wnt pathway was analysed by a reporter gene assay. RESULTS Besides pro-calcifying culture conditions, also activation of Wnt signaling by a specific agonist (under normal culture conditions) stimulated VSMC-calcification accompanied by enhanced expression and secretion of the gelatinases MMP-2 and -9. Vice versa, recombinant MMP-2 and -9 induced a time-delayed activation of Wnt signaling after 72 h in VSMC but showed no direct effects after 24-48 h. These effects were blocked by pharmacological inhibition of MMPs or of Wnt signaling. CONCLUSIONS Our study suggests that the pro-calcifying environment in CKD induces Wnt signaling in VSMC which in turn contributes to the induction of MMPs which then foster the development of arteriosclerosis. Thus, besides MMP inhibition, the inhibition of Wnt signaling in VSMC might represent a therapeutic target for the prevention of vascular calcifications.
Collapse
Affiliation(s)
- Christian Freise
- Center for Cardiovascular Research, Charité - University Medicine, Campus Mitte, Hessische Str. 3-4, 10115, Berlin, Germany. .,Department of Pediatric Nephrology, Charité - University Medicine, Campus Virchow Clinic, 13353, Berlin, Germany.
| | - Nadja Kretzschmar
- Center for Cardiovascular Research, Charité - University Medicine, Campus Mitte, Hessische Str. 3-4, 10115, Berlin, Germany.,Department of Pediatric Nephrology, Charité - University Medicine, Campus Virchow Clinic, 13353, Berlin, Germany
| | - Uwe Querfeld
- Center for Cardiovascular Research, Charité - University Medicine, Campus Mitte, Hessische Str. 3-4, 10115, Berlin, Germany.,Department of Pediatric Nephrology, Charité - University Medicine, Campus Virchow Clinic, 13353, Berlin, Germany
| |
Collapse
|
157
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
158
|
Real-Time Analysis of Endogenous Wnt Signalling in 3D Mesenchymal Stromal Cells. Stem Cells Int 2016; 2016:7132529. [PMID: 27668000 PMCID: PMC5030414 DOI: 10.1155/2016/7132529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/08/2016] [Indexed: 01/08/2023] Open
Abstract
Wnt signalling has been implicated in the regulation of stem cell self-renewal and differentiation; however, the majority of in vitro studies are carried out using monolayer 2D culture techniques. Here, we used mesenchymal stromal cell (MSC) EGFP reporter lines responsive to Wnt pathway activation in a 3D spheroid culture system to mimic better the in vivo environment. Endogenous Wnt signalling was then investigated under basal conditions and when MSCs were induced to undergo osteogenic and adipogenic differentiation. Interestingly, endogenous Wnt signalling was only active during 3D differentiation whereas 2D cultures showed no EGFP expression throughout an extended differentiation time-course. Furthermore, exogenous Wnt signalling in 3D adipogenic conditions inhibited differentiation compared to unstimulated controls. In addition, suppressing Wnt signalling by Dkk-1 restored and facilitated adipogenic differentiation in MSC spheroids. Our findings indicate that endogenous Wnt signalling is active and can be tracked in 3D MSC cultures where it may act as a molecular switch in adipogenesis. The identification of the signalling pathways that regulate MSCs in a 3D in vivo-like environment will advance our understanding of the molecular mechanisms that control MSC fate.
Collapse
|
159
|
Characterization of Cellular and Molecular Heterogeneity of Bone Marrow Stromal Cells. Stem Cells Int 2016; 2016:9378081. [PMID: 27610142 PMCID: PMC5004045 DOI: 10.1155/2016/9378081] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/26/2016] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived stromal stem cells (hBMSC) exhibit multiple functions, including differentiation into skeletal cells (progenitor function), hematopoiesis support, and immune regulation (nonprogenitor function). We have previously demonstrated the presence of morphological and functional heterogeneity of hBMSC cultures. In the present study, we characterized in detail two hTERT-BMSC clonal cell populations termed here CL1 and CL2 that represent an opposing phenotype with respect to morphology, markers expression: alkaline phosphatase (ALP) and CD146, and ex vivo differentiation potential. CL1 differentiated readily to osteoblasts, adipocytes, and chondrocytes as shown by expression of lineage specific genes and proteins. Whole genome transcriptome profiling of CL1 versus CL2 revealed enrichment in CL1 of bone-, mineralization-, and skeletal muscle-related genes, for example, ALP, POSTN, IGFBP5 BMP4, and CXCL12. On the other hand, CL2 transcriptome was enriched in immune modulatory genes, for example, CD14, CD99, NOTCH3, CXCL6, CFB, and CFI. Furthermore, gene expression microarray analysis of osteoblast differentiated CL1 versus CL2 showed significant upregulation in CL1 of bone development and osteoblast differentiation genes which included several homeobox genes: TBX15, HOXA2 and HOXA10, and IGF1, FGFR3, BMP6, MCAM, ITGA10, IGFBP5, and ALP. siRNA-based downregulation of the ALP gene in CL1 impaired osteoblastic and adipocytic differentiation. Our studies demonstrate the existence of molecular and functional heterogeneity in cultured hBMSC. ALP can be employed to identify osteoblastic and adipocytic progenitor cells in the heterogeneous hBMSC cultures.
Collapse
|
160
|
Phetfong J, Sanvoranart T, Nartprayut K, Nimsanor N, Seenprachawong K, Prachayasittikul V, Supokawej A. Osteoporosis: the current status of mesenchymal stem cell-based therapy. Cell Mol Biol Lett 2016; 21:12. [PMID: 28536615 PMCID: PMC5414670 DOI: 10.1186/s11658-016-0013-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/25/2016] [Indexed: 12/21/2022] Open
Abstract
Osteoporosis, or bone loss, is a progressive, systemic skeletal disease that affects millions of people worldwide. Osteoporosis is generally age related, and it is underdiagnosed because it remains asymptomatic for several years until the development of fractures that confine daily life activities, particularly in elderly people. Most patients with osteoporotic fractures become bedridden and are in a life-threatening state. The consequences of fracture can be devastating, leading to substantial morbidity and mortality of the patients. The normal physiologic process of bone remodeling involves a balance between bone resorption and bone formation during early adulthood. In osteoporosis, this process becomes imbalanced, resulting in gradual losses of bone mass and density due to enhanced bone resorption and/or inadequate bone formation. Several growth factors underlying age-related osteoporosis and their signaling pathways have been identified, such as osteoprotegerin (OPG)/receptor activator of nuclear factor B (RANK)/RANK ligand (RANKL), bone morphogenetic protein (BMP), wingless-type MMTV integration site family (Wnt) proteins and signaling through parathyroid hormone receptors. In addition, the pathogenesis of osteoporosis has been connected to genetics. The current treatment of osteoporosis predominantly consists of antiresorptive and anabolic agents; however, the serious adverse effects of using these drugs are of concern. Cell-based replacement therapy via the use of mesenchymal stem cells (MSCs) may become one of the strategies for osteoporosis treatment in the future.
Collapse
Affiliation(s)
- Jitrada Phetfong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Tanwarat Sanvoranart
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Kuneerat Nartprayut
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Natakarn Nimsanor
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Kanokwan Seenprachawong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| |
Collapse
|
161
|
Van Camp JK, Beckers S, Zegers D, Verhulst SL, Van Hoorenbeeck K, Massa G, Verrijken A, Desager KN, Van Gaal LF, Van Hul W. Nucleotide variation of sFRP5 gene is not associated with obesity in children and adolescents. Mol Biol Rep 2016; 43:1041-7. [PMID: 27497818 DOI: 10.1007/s11033-016-4050-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/29/2016] [Indexed: 12/12/2022]
Abstract
Because sFRP5 was shown to be an important extracellular modulator of the Wnt pathway, regulating adipogenesis, we wanted to investigate the role of sFRP5 variants in human, monogenic obesity by performing mutation analysis. We screened the complete sFRP5 coding region in 622 obese children and adolescents and 503 lean control individuals by high-resolution melting curve analysis and direct sequencing. We found a total of 15 sequence variants in sFRP5, 10 of which resulted in a non-synonymous amino acid change. Five of these variants were, to our knowledge, not previously reported. For one of the variants (c.-3G>A), we identified a trend towards association between the variant frequency and the obese phenotype. We argue that, when looking at conservation and location inside known protein domains, several of the identified variants (D103N, A113V, K212N and H317L), may affect sFRP5 protein function. In addition, we found c.-3G>A, residing in the Kozak sequence, with a lower frequency in cases compared to controls. However, functional studies investigating the effect of sFRP5 variants on protein function are necessary to determine the true role of sFRP5 genetic variation in human, monogenic obesity.
Collapse
Affiliation(s)
- Jasmijn K Van Camp
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Sigri Beckers
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Doreen Zegers
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Stijn L Verhulst
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Kim Van Hoorenbeeck
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Guy Massa
- Department of Paediatrics, Jessa Hospital, Stadsomvaart 11, 3500, Hasselt, Belgium
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Kristine N Desager
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Luc F Van Gaal
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.
| |
Collapse
|
162
|
Abstract
Recent understanding of the cellular and molecular signaling activations in adult mesenchymal stem cells (MSCs) has provided new insights into their potential clinical applications, particularly for tissue repair and regeneration. This review focuses on these advances, specifically in the context of self-renewal for tissue repair and recovery after radiation injury. Thus far, MSCs have been characterized extensively and shown to be useful in mitigation and therapy for acute radiation syndrome and cognitive dysfunction. Use of MSCs for treating radiation injury alone or in combination with additional trauma is foreseeable.
Collapse
Affiliation(s)
- Juliann G Kiang
- *Scientific Research Department, Armed Forces Radiobiology Research Institute; Department of Radiation Biology, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20889-1076
| |
Collapse
|
163
|
Thiele S, Rachner TD, Rauner M, Hofbauer LC. WNT5A and Its Receptors in the Bone-Cancer Dialogue. J Bone Miner Res 2016; 31:1488-96. [PMID: 27355180 DOI: 10.1002/jbmr.2899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/08/2016] [Accepted: 06/24/2016] [Indexed: 12/12/2022]
Abstract
Wnt signaling is critical for tumorigenesis and skeletal remodeling. However, its contribution to the formation of metastatic bone lesions remains poorly defined. One major challenge of unraveling its role in cancer progression is the high complexity of Wnt signaling, which includes numerous ligands, receptors, and inhibitors, with intricate biological effects and specific signaling pathways depending on the cellular context. In this perspective, we summarize the role of the noncanonical Wnt ligand WNT5A in the development and metastatic process of osteotropic cancer entities. We focus on its tumor-suppressive function in breast cancer, tumor promoting effects in melanoma, and ambiguous role in prostate cancer, and discuss potential challenges and opportunities that may be associated with targeting Wnt signaling for cancer therapy and treatment of bone metastases. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stefanie Thiele
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Tilman D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität (TU) Dresden Medical Center, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Healthy Aging, Technische Universität (TU) Dresden Medical Center, Dresden, Germany
| |
Collapse
|
164
|
MicroRNAs in regulation of osteogenic differentiation of mesenchymal stem cells. Cell Tissue Res 2016; 368:229-238. [DOI: 10.1007/s00441-016-2462-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022]
|
165
|
Articular cartilage repair: Current needs, methods and research directions. Semin Cell Dev Biol 2016; 62:67-77. [PMID: 27422331 DOI: 10.1016/j.semcdb.2016.07.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022]
Abstract
Articular cartilage is a highly specialized tissue whose remarkable properties of deformability, resistance to mechanical loading, and low-friction gliding are essential to joint function. Due to its role as a cushion in bone articulation, articular cartilage is subject to many types of damaging insults, including decades of wear and tear, and acute joint injuries. However, this built-for-life tissue has a very poor intrinsic ability in adulthood to durably heal defects created by damaging insults. Consequently, articular cartilage progressively deteriorates and is eventually eroded, exposing the subchondral bone to the joint space, triggering inflammation and osteophyte development, and generating severe pain and joint incapacitation. The disease is called osteoarthritis (OA) and is today the leading cause of pain and disability in the human population. Researchers and clinicians have worked for decades to develop strategies to treat OA and restore joint function, but they are still far from being able to offer patients effective preventive or restorative treatments. Novel ideas, knowledge and technologies that nurture hope for major new breakthroughs are therefore sought. In this review, we first outline the composition, structure, and functional properties of normal human adult articular cartilage, as a reference for tissue conservation and regenerative strategies. We then describe current options that have been used clinically and in pre-clinical trials to treat osteoarthritic patients, and we discuss the benefits and inadequacies of these treatment options. Next, we review research efforts that are currently ongoing to try and achieve durable repair of functional cartilage tissue. Methods include engineering of tissue implants and we discuss the needs and options for tissue scaffolds, cell sources, and growth and differentiation factors to generate de novo or repair bona fide articular cartilage.
Collapse
|
166
|
Lowndes M, Rotherham M, Price JC, El Haj AJ, Habib SJ. Immobilized WNT Proteins Act as a Stem Cell Niche for Tissue Engineering. Stem Cell Reports 2016; 7:126-37. [PMID: 27411105 PMCID: PMC4944585 DOI: 10.1016/j.stemcr.2016.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 11/18/2022] Open
Abstract
The timing, location, and level of WNT signaling are highly regulated during embryonic development and for the maintenance of adult tissues. Consequently the ability to provide a defined and directed source of WNT proteins is crucial to fully understand its role in tissue development and to mimic its activity in vitro. Here we describe a one-step immobilization technique to covalently bind WNT3A proteins as a basal surface with easy storage and long-lasting activity. We show that this platform is able to maintain adult and embryonic stem cells while also being adaptable for 3D systems. Therefore, this platform could be used for recapitulating specific stem cell niches with the goal of improving tissue engineering.
Collapse
Affiliation(s)
- Molly Lowndes
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Michael Rotherham
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Stoke-on-Trent ST4 7QB, UK
| | - Joshua C Price
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK; Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Stoke-on-Trent ST4 7QB, UK
| | - Alicia J El Haj
- Institute for Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Stoke-on-Trent ST4 7QB, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK.
| |
Collapse
|
167
|
Collette NM, Yee CS, Hum NR, Murugesh DK, Christiansen BA, Xie L, Economides AN, Manilay JO, Robling AG, Loots GG. Sostdc1 deficiency accelerates fracture healing by promoting the expansion of periosteal mesenchymal stem cells. Bone 2016; 88:20-30. [PMID: 27102547 PMCID: PMC6277141 DOI: 10.1016/j.bone.2016.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/16/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Loss of Sostdc1, a growth factor paralogous to Sost, causes the formation of ectopic incisors, fused molars, abnormal hair follicles, and resistance to kidney disease. Sostdc1 is expressed in the periosteum, a source of osteoblasts, fibroblasts and mesenchymal progenitor cells, which are critically important for fracture repair. Here, we investigated the role of Sostdc1 in bone metabolism and fracture repair. Mice lacking Sostdc1 (Sostdc1(-/-)) had a low bone mass phenotype associated with loss of trabecular bone in both lumbar vertebrae and in the appendicular skeleton. In contrast, Sostdc1(-/-) cortical bone measurements revealed larger bones with higher BMD, suggesting that Sostdc1 exerts differential effects on cortical and trabecular bone. Mid-diaphyseal femoral fractures induced in Sostdc1(-/-) mice showed that the periosteal population normally positive for Sostdc1 rapidly expands during periosteal thickening and these cells migrate into the fracture callus at 3days post fracture. Quantitative analysis of mesenchymal stem cell (MSC) and osteoblast populations determined that MSCs express Sostdc1, and that Sostdc1(-/-) 5day calluses harbor >2-fold more MSCs than fractured wildtype controls. Histologically a fraction of Sostdc1-positive cells also expressed nestin and α-smooth muscle actin, suggesting that Sostdc1 marks a population of osteochondral progenitor cells that actively participate in callus formation and bone repair. Elevated numbers of MSCs in D5 calluses resulted in a larger, more vascularized cartilage callus at day 7, and a more rapid turnover of cartilage with significantly more remodeled bone and a thicker cortical shell at 21days post fracture. These data support accelerated or enhanced bone formation/remodeling of the callus in Sostdc1(-/-) mice, suggesting that Sostdc1 may promote and maintain mesenchymal stem cell quiescence in the periosteum.
Collapse
Affiliation(s)
- Nicole M Collette
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | - Cristal S Yee
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA; Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA
| | - Nicholas R Hum
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | - Deepa K Murugesh
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | | | - LiQin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Jennifer O Manilay
- Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA
| | | | - Gabriela G Loots
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA; Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA.
| |
Collapse
|
168
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
169
|
Keller KC, Ding H, Tieu R, Sparks NRL, Ehnes DD, Zur Nieden NI. Wnt5a Supports Osteogenic Lineage Decisions in Embryonic Stem Cells. Stem Cells Dev 2016; 25:1020-32. [PMID: 26956615 DOI: 10.1089/scd.2015.0367] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The specification of pluripotent stem cells into the bone-forming osteoblasts has been explored in a number of studies. However, the current body of literature has yet to adequately address the role of Wnt glycoproteins in the differentiation of pluripotent stem cells along the osteogenic lineage. During mouse embryonic stem cell (ESC) in vitro osteogenesis, the noncanonical WNT5a is expressed early on. Cells either sorted by their positive WNT5a expression or when supplemented with recombinant WNT5a (rWNT5a) during a 2-day window showed significantly enhanced osteogenic yield. Mechanistically, rWNT5a supplementation upregulated protein kinase C (PKC), calcium/calmodulin-dependent kinase II (CamKII) and c-Jun N-terminal kinase (JNK) activity while antagonizing the key effector of canonical Wnt signaling: β-catenin. Conversely, when recombinant WNT3a (rWNT3a) or other positive regulators of β-catenin were employed during this same time window there was a decrease in osteogenic marker expression. However, if rWNT3a was supplemented during a time window following rWNT5a treatment, osteogenic differentiation was enhanced both in murine and human ESCs. Elucidating the role of these WNT ligands in directing the early stages of osteogenesis has the potential to considerably improve tissue engineering protocols and applications for regenerative medicine.
Collapse
Affiliation(s)
- Kevin C Keller
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Huawen Ding
- 2 Applied Stem Cell Technologies Unit, Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| | - Rudy Tieu
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Nicole R L Sparks
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Devon D Ehnes
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Nicole I Zur Nieden
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California.,2 Applied Stem Cell Technologies Unit, Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| |
Collapse
|
170
|
Becker M, Potapenko T, Niklaus A, Bieback K, Ho AD, Müller AM. Polycomb Protein BMI1 Regulates Osteogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells Downstream of GSK3. Stem Cells Dev 2016; 25:922-33. [PMID: 27100571 DOI: 10.1089/scd.2015.0277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Polycomb proteins such as the B lymphoma Mo-MLV insertion region 1 homolog (BMI1) are essential chromatin factors for the self-renewal and differentiation of embryonic and adult stem cells. BMI1 also plays a critical role in osteogenesis as Bmi1-deficient mice display a skeletal phenotype caused by the exhaustion of the mesenchymal stem cell pool. In this study, we have studied the role of BMI1 in the osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs). BMI1 protein, but not RNA levels, increases during in vitro osteogenic differentiation of hASCs. Overexpression of BMI1 leads to an osteogenic priming of hASCs under nondifferentiating conditions and enhanced osteogenesis upon differentiation, along with increased BMP2 and WNT11 expressions. Conversely, knockdown of BMI1 expression reduces osteogenic differentiation. Furthermore, our studies indicate that during osteogenic differentiation of hASCs, BMI1 is a downstream target of GSK3 signaling. BMI1, therefore, acts as a pro-osteogenic differentiation factor in hASCs and hence it is a promising target for active modulation of hASC-derived osteogenesis.
Collapse
Affiliation(s)
- Matthias Becker
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Tamara Potapenko
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Andrea Niklaus
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Karen Bieback
- 2 Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg , German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Anthony D Ho
- 3 Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Albrecht M Müller
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| |
Collapse
|
171
|
Ali D, Hamam R, Alfayez M, Kassem M, Aldahmash A, Alajez NM. Epigenetic Library Screen Identifies Abexinostat as Novel Regulator of Adipocytic and Osteoblastic Differentiation of Human Skeletal (Mesenchymal) Stem Cells. Stem Cells Transl Med 2016; 5:1036-47. [PMID: 27194745 DOI: 10.5966/sctm.2015-0331] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/10/2016] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : The epigenetic mechanisms promoting lineage-specific commitment of human skeletal (mesenchymal or stromal) stem cells (hMSCs) into adipocytes or osteoblasts are still not fully understood. Herein, we performed an epigenetic library functional screen and identified several novel compounds, including abexinostat, which promoted adipocytic and osteoblastic differentiation of hMSCs. Using gene expression microarrays, chromatin immunoprecipitation for H3K9Ac combined with high-throughput DNA sequencing (ChIP-seq), and bioinformatics, we identified several key genes involved in regulating stem cell proliferation and differentiation that were targeted by abexinostat. Concordantly, ChIP-quantitative polymerase chain reaction revealed marked increase in H3K9Ac epigenetic mark on the promoter region of AdipoQ, FABP4, PPARγ, KLF15, CEBPA, SP7, and ALPL in abexinostat-treated hMSCs. Pharmacological inhibition of focal adhesion kinase (PF-573228) or insulin-like growth factor-1R/insulin receptor (NVP-AEW51) signaling exhibited significant inhibition of abexinostat-mediated adipocytic differentiation, whereas inhibition of WNT (XAV939) or transforming growth factor-β (SB505124) signaling abrogated abexinostat-mediated osteogenic differentiation of hMSCs. Our findings provide insight into the understanding of the relationship between the epigenetic effect of histone deacetylase inhibitors, transcription factors, and differentiation pathways governing adipocyte and osteoblast differentiation. Manipulating such pathways allows a novel use for epigenetic compounds in hMSC-based therapies and tissue engineering. SIGNIFICANCE This unbiased epigenetic library functional screen identified several novel compounds, including abexinostat, that promoted adipocytic and osteoblastic differentiation of human skeletal (mesenchymal or stromal) stem cells (hMSCs). These data provide new insight into the understanding of the relationship between the epigenetic effect of histone deacetylase inhibitors, transcription factors, and differentiation pathways controlling adipocyte and osteoblast differentiation of hMSCs. Manipulating such pathways allows a novel use for epigenetic compounds in hMSC-based therapies for tissue engineering, bone disease, obesity, and metabolic-disorders.
Collapse
Affiliation(s)
- Dalia Ali
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Rimi Hamam
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Musaed Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Moustapha Kassem
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia Molecular Endocrinology and Stem Cell Research Unit, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia Prince Naif Health Research Center, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Nehad M Alajez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
172
|
Rios C, D'Ippolito G, Curtis KM, Delcroix GJR, Gomez LA, El Hokayem J, Rieger M, Parrondo R, de Las Pozas A, Perez-Stable C, Howard GA, Schiller PC. Low Oxygen Modulates Multiple Signaling Pathways, Increasing Self-Renewal, While Decreasing Differentiation, Senescence, and Apoptosis in Stromal MIAMI Cells. Stem Cells Dev 2016; 25:848-60. [PMID: 27059084 DOI: 10.1089/scd.2015.0362] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human bone marrow multipotent mesenchymal stromal cell (hMSC) number decreases with aging. Subpopulations of hMSCs can differentiate into cells found in bone, vasculature, cartilage, gut, and other tissues and participate in their repair. Maintaining throughout adult life such cell subpopulations should help prevent or delay the onset of age-related degenerative conditions. Low oxygen tension, the physiological environment in progenitor cell-rich regions of the bone marrow microarchitecture, stimulates the self-renewal of marrow-isolated adult multilineage inducible (MIAMI) cells and expression of Sox2, Nanog, Oct4a nuclear accumulation, Notch intracellular domain, notch target genes, neuronal transcriptional repressor element 1 (RE1)-silencing transcription factor (REST), and hypoxia-inducible factor-1 alpha (HIF-1α), and additionally, by decreasing the expression of (i) the proapoptotic proteins, apoptosis-inducing factor (AIF) and Bak, and (ii) senescence-associated p53 expression and β-galactosidase activity. Furthermore, low oxygen increases canonical Wnt pathway signaling coreceptor Lrp5 expression, and PI3K/Akt pathway activation. Lrp5 inhibition decreases self-renewal marker Sox2 mRNA, Oct4a nuclear accumulation, and cell numbers. Wortmannin-mediated PI3K/Akt pathway inhibition leads to increased osteoblastic differentiation at both low and high oxygen tension. We demonstrate that low oxygen stimulates a complex signaling network involving PI3K/Akt, Notch, and canonical Wnt pathways, which mediate the observed increase in nuclear Oct4a and REST, with simultaneous decrease in p53, AIF, and Bak. Collectively, these pathway activations contribute to increased self-renewal with concomitant decreased differentiation, cell cycle arrest, apoptosis, and/or senescence in MIAMI cells. Importantly, the PI3K/Akt pathway plays a central mechanistic role in the oxygen tension-regulated self-renewal versus osteoblastic differentiation of progenitor cells.
Collapse
Affiliation(s)
- Carmen Rios
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida.,2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida
| | - Gianluca D'Ippolito
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida.,3 Department of Orthopaedics, University of Miami Miller School of Medicine , Miami, Florida.,4 Geriatrics Institute, University of Miami Miller School of Medicine , Miami, Florida.,5 Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida.,6 Department of Biomedical Engineering, University of Miami , Coral Gables, Florida
| | - Kevin M Curtis
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida.,2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida
| | - Gaëtan J-R Delcroix
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida.,3 Department of Orthopaedics, University of Miami Miller School of Medicine , Miami, Florida.,5 Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida
| | - Lourdes A Gomez
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida
| | - Jimmy El Hokayem
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida
| | - Megan Rieger
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida
| | - Ricardo Parrondo
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida
| | - Alicia de Las Pozas
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida
| | - Carlos Perez-Stable
- 2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida.,7 Department of Medicine, University of Miami Miller School of Medicine , Miami, Florida
| | - Guy A Howard
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida.,2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida.,7 Department of Medicine, University of Miami Miller School of Medicine , Miami, Florida
| | - Paul C Schiller
- 1 Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, Florida.,2 GRECC and Research Service, Veterans Affairs Medical Center , Miami, Florida.,3 Department of Orthopaedics, University of Miami Miller School of Medicine , Miami, Florida.,4 Geriatrics Institute, University of Miami Miller School of Medicine , Miami, Florida.,5 Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
173
|
Belinsky GS, Sreekumar B, Andrejecsk JW, Saltzman WM, Gong J, Herzog RI, Lin S, Horsley V, Carpenter TO, Chung C. Pigment epithelium-derived factor restoration increases bone mass and improves bone plasticity in a model of osteogenesis imperfecta type VI via Wnt3a blockade. FASEB J 2016; 30:2837-48. [PMID: 27127101 DOI: 10.1096/fj.201500027r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 04/18/2016] [Indexed: 12/23/2022]
Abstract
Null mutations in for pigment epithelium-derived factor (PEDF), the protein product of the SERPINF1 gene, are the cause of osteogenesis imperfecta (OI) type VI. The PEDF-knockout (KO) mouse captures crucial elements of the human disease, including diminished bone mineralization and propensity to fracture. Our group and others have demonstrated that PEDF directs human mesenchymal stem cell (hMSC) commitment to the osteoblast lineage and modulates Wnt/β-catenin signaling, a major regulator of bone development; however, the ability of PEDF to restore bone mass in a mouse model of OI type VI has not been determined. In this study, PEDF delivery increased trabecular bone volume/total volume by 52% in 6-mo-old PEDF-KO mice but not in wild-type mice. In young (19-d-old) PEDF-KO mice, PEDF restoration increased bone volume fraction by 35% and enhanced biomechanical parameters of bone plasticity. A Wnt-green fluorescent protein reporter demonstrated dynamic changes in Wnt/β-catenin signaling characterized by early activation and marked suppression during terminal differentiation of hMSCs. Continuous Wnt3a exposure impeded mineralization of hMSCs, whereas the combination of Wnt3a and PEDF potentiated mineralization. Interrogation of the PEDF sequence identified a conserved motif found in other Wnt modulators, such as the dickkopf proteins. Mutation of a single amino acid on a 34-mer PEDF peptide increased mineralization of hMSC cultures compared with the native peptide sequence. These results indicate that PEDF counters Wnt signaling to allow for osteoblast differentiation and provides a mechanistic insight into how the PEDF null state results in OI type VI.-Belinsky, G. S., Sreekumar, B., Andrejecsk, J. W., Saltzman, W. M., Gong, J., Herzog, R. I., Lin, S., Horsley, V., Carpenter, T. O., Chung, C. Pigment epithelium-derived factor restoration increases bone mass and improves bone plasticity in a model of osteogenesis imperfecta type VI via Wnt3a blockade.
Collapse
Affiliation(s)
- Glenn S Belinsky
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bharath Sreekumar
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jillian W Andrejecsk
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Jingjing Gong
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Raimund I Herzog
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Samantha Lin
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Thomas O Carpenter
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
174
|
Yu G, Wang J, Lin X, Diao S, Cao Y, Dong R, Wang L, Wang S, Fan Z. Demethylation of SFRP2 by histone demethylase KDM2A regulated osteo-/dentinogenic differentiation of stem cells of the apical papilla. Cell Prolif 2016; 49:330-40. [PMID: 27074224 DOI: 10.1111/cpr.12256] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/08/2016] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Dental mesenchymal stem cells (MSCs) are easily obtained; however, mechanisms underlying directed differentiation of these cells remains unclear. Wnt/β-catenin signalling is essential for mesenchymal cell commitment and differentiation, and Wnt inhibition is linked to stem cell maintenance and function. Secreted frizzled-related protein 2 (SFRP2) competes with the Frizzled receptor for direct binding to Wnt and blocks activation of Wnt signalling. Here, we used stem cells derived from apical papillae (SCAPs) to study the functions of SFRP2. MATERIALS AND METHODS SCAPs were isolated from apical papillae of immature third molars. The cells were analysed using alkaline phosphatase activity assays, Alizarin red staining and quantitative calcium measurements. In addition, we evaluated expression profile of genes associated with osteogenesis and dentinogenesis (osteo-/dentinogenesis), and conducted in vivo transplantation experiments to determine osteo-/dentinogenic differentiation potential of SCAPs. ChIP assays were used to detect histone methylation at the SFRP2 promoter. RESULTS We found that SFRP2 enhanced osteo-/dentinogenic differentiation via Osterix, a key transcription factor in SCAPs. Furthermore, silencing SFRP2 induced SCAP cell death in osteogenic-inducing medium, indicating that SFRP2 is a key factor in maintaining SCAP survival following osteo-/dentinogenic commitment. Moreover, we found that silencing KDM2A, a histone demethylase and BCL6 co-repressor, de-repressed SFRP2 transcription by increasing histone H3K4 and H3K36 methylation at the SFRP2 promoter. CONCLUSIONS Our results have identified a new function of SFRP2 and shed new light on the molecular mechanism underlying directed differentiation of stem cells of dental origin.
Collapse
Affiliation(s)
- Guoxia Yu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Stomatology, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Jinsong Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Implant Dentistry, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yu Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| |
Collapse
|
175
|
Shi C, Lv T, Xiang Z, Sun Z, Qian W, Han X. Role of Wnt/β-Catenin Signaling in Epithelial Differentiation of Lung Resident Mesenchymal Stem Cells. J Cell Biochem 2016; 116:1532-9. [PMID: 25546504 DOI: 10.1002/jcb.25069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/18/2014] [Indexed: 12/22/2022]
Abstract
Accumulating evidence has demonstrated that stem cells have the ability to repair the lung tissue injuries following either injection of cultured cells or bone marrow transplantation. As a result, increasing attention has focused on the lung resident mesenchymal stem cells (LR-MSCs) for repairing damaged lung tissues. Meanwhile, some studies have revealed that Wnt/β-catenin signaling plays an important role in the epithelial differentiation of mesenchymal stem cells (MSCs). In the current study, our aim was to explore the roles of Wnt/β-catenin signaling on cell proliferation and epithelial differentiation of LR-MSCs. We have successfully isolated the stem cell antigen (Sca)-1(+) CD45(-) CD31(-) cells which were proposed to be LR-MSCs by magnetic-activated cell sorting (MACS). Furthermore, we demonstrated the expression of epithelial markers on LR-MSCs following indirect co-culture of these cells with alveolar epithelial type II (ATII) cells, confirming the epithelial phenotype of LR-MSCs following co-culture. In order to clarify the regulatory mechanisms of Wnt/β-catenin signaling in epithelial differentiation of LR-MSCs, we measured the protein levels of several important members involved in Wnt/β-catenin signaling in the presence or absence of some canonical activators and inhibitors of the β-catenin pathways. In conclusion, our study demonstrated that Wnt/β-catenin signaling may be an essential mechanism underlying the regulation of epithelial differentiation of LR-MSCs.
Collapse
Affiliation(s)
- Chaowen Shi
- Immunology and Reproductive Biology Laboratory, Medical School of Nanjing University, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Tengfei Lv
- Immunology and Reproductive Biology Laboratory, Medical School of Nanjing University, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Zou Xiang
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Zhaorui Sun
- Department of Emergency, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, PR China
| | - Weiping Qian
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210093, China
| | - Xiaodong Han
- Immunology and Reproductive Biology Laboratory, Medical School of Nanjing University, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, 210093, China
| |
Collapse
|
176
|
Moschouris P, Retzepi M, Petrie A, Donos N. Effect of Wnt3a delivery on early healing events during guided bone regeneration. Clin Oral Implants Res 2016; 28:283-290. [DOI: 10.1111/clr.12796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2016] [Indexed: 12/21/2022]
Affiliation(s)
- P Moschouris
- Periodontology Unit; Department of Clinical Research; UCL Eastman Dental Institute; London UK
| | - M Retzepi
- Periodontology Unit; Department of Clinical Research; UCL Eastman Dental Institute; London UK
| | - A Petrie
- Biostatistics Unit; UCL Eastman Dental Institute; London UK
| | - N Donos
- Periodontology Unit; Department of Clinical Research; UCL Eastman Dental Institute; London UK
- Centre for Oral Clinical Research; Institute of Dentistry; Barts & The London School of Medicine & Dentistry; London UK
| |
Collapse
|
177
|
TLR4 Activation Promotes Bone Marrow MSC Proliferation and Osteogenic Differentiation via Wnt3a and Wnt5a Signaling. PLoS One 2016; 11:e0149876. [PMID: 26930594 PMCID: PMC4773221 DOI: 10.1371/journal.pone.0149876] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/06/2016] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from adult bone marrow maintain their self-renewal ability and the ability to differentiate into osteoblast. Thus, adult bone marrow MSCs play a key role in the regeneration of bone tissue. Previous studies indicated that TLR4 is expressed in MSCs and is critical in regulating the fate decision of MSCs. However, the exact functional role and underlying mechanisms of how TLR4 regulate bone marrow MSC proliferation and differentiation are unclear. Here, we found that activated TLR4 by its ligand LPS promoted the proliferation and osteogenic differentiation of MSCs in vitro. TLR4 activation by LPS also increased cytokine IL-6 and IL-1β production in MSCs. In addition, LPS treatment has no effect on inducing cell death of MSCs. Deletion of TLR4 expression in MSCs completely eliminated the effects of LPS on MSC proliferation, osteogenic differentiation and cytokine production. We also found that the mRNA and protein expression of Wnt3a and Wnt5a, two important factors in regulating MSC fate decision, was upregulated in a TLR4-dependent manner. Silencing Wnt3a with specific siRNA remarkably inhibited TLR4-induced MSC proliferation, while Wnt5a specific siRNA treatment significantly antagonized TLR4-induced MSC osteogenic differentiation. These results together suggested that TLR4 regulates bone marrow MSC proliferation and osteogenic differentiation through Wnt3a and Wnt5a signaling. These finding provide new data to understand the role and the molecular mechanisms of TLR4 in regulating bone marrow MSC functions. These data also provide new insight in developing new therapy in bone regeneration using MSCs by modulating TLR4 and Wnt signaling activity.
Collapse
|
178
|
MATSUNAGA K, ITO C, NAKAKOGAWA K, SUGIUCHI A, SAKO R, FURUSAWA M, MURAMATSU T. Response to light compressive force in human cementoblasts in vitro . Biomed Res 2016; 37:293-298. [DOI: 10.2220/biomedres.37.293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Kenji MATSUNAGA
- Department of Endodontics and Clinical Cariology, Tokyo Dental College
| | - Chika ITO
- Department of Oral Health Science Center, Tokyo Dental College
| | | | - Akina SUGIUCHI
- Department of Endodontics and Clinical Cariology, Tokyo Dental College
| | - Ryo SAKO
- Department of Endodontics and Clinical Cariology, Tokyo Dental College
| | - Masahiro FURUSAWA
- Department of Endodontics and Clinical Cariology, Tokyo Dental College
| | - Takashi MURAMATSU
- Department of Endodontics and Clinical Cariology, Tokyo Dental College
| |
Collapse
|
179
|
Falconi G, Fabiani E, Fianchi L, Criscuolo M, Raffaelli CS, Bellesi S, Hohaus S, Voso MT, D’Alò F, Leone G. Impairment of PI3K/AKT and WNT/β-catenin pathways in bone marrow mesenchymal stem cells isolated from patients with myelodysplastic syndromes. Exp Hematol 2016; 44:75-83.e1-4. [DOI: 10.1016/j.exphem.2015.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/14/2015] [Accepted: 10/19/2015] [Indexed: 01/01/2023]
|
180
|
Bischoff DS, Zhu JH, Makhijani NS, Yamaguchi DT. Induction of CXC chemokines in human mesenchymal stem cells by stimulation with secreted frizzled-related proteins through non-canonical Wnt signaling. World J Stem Cells 2015; 7:1262-1273. [PMID: 26730270 PMCID: PMC4691694 DOI: 10.4252/wjsc.v7.i11.1262] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/03/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of secreted frizzled-related proteins (sFRPs) on CXC chemokine expression in human mesenchymal stem cells (hMSCs).
METHODS: CXC chemokines such as CXCL5 and CXCL8 are induced in hMSCs during differentiation with osteogenic differentiation medium (OGM) and may be involved in angiogenic stimulation during bone repair. hMSCs were treated with conditioned medium (CM) from L-cells expressing non-canonical Wnt5a protein, or with control CM from wild type L-cells, or directly with sFRPs for up to 10 d in culture. mRNA expression levels of both CXCL5 and CXCL8 were quantitated by real-time reverse transcriptase-polymerase chain reaction and secreted protein levels of these proteins determined by ELISA. Dose- (0-500 ng/mL) and time-response curves were generated for treatment with sFRP1. Signal transduction pathways were explored by western blot analysis with pan- or phosphorylation-specific antibodies, through use of specific pathway inhibitors, and through use of siRNAs targeting specific frizzled receptors (Fzd)-2 and 5 or the receptor tyrosine kinase-like orphan receptor-2 (RoR2) prior to treatment with sFRPs.
RESULTS: CM from L-cells expressing Wnt5a, a non-canonical Wnt, stimulated an increase in CXCL5 mRNA expression and protein secretion in comparison to control L-cell CM. sFRP1, which should inhibit both canonical and non-canonical Wnt signaling, surprisingly enhanced the expression of CXCL5 at 7 and 10 d. Dickkopf1, an inhibitor of canonical Wnt signaling prevented the sFRP-stimulated induction of CXCL5 and actually inhibited basal levels of CXCL5 expression at 7 but not at 10 d post treatment. In addition, all four sFRPs isoforms induced CXCL8 expression in a dose- and time-dependent manner with maximum expression at 7 d with treatment at 150 ng/mL. The largest increases in CXCL5 expression were seen from stimulation with sFRP1 or sFRP2. Analysis of mitogen-activated protein kinase signaling pathways in the presence of OGM showed sFRP1-induced phosphorylation of extracellular signal-regulated kinase (ERK) (p44/42) maximally at 5 min after sFRP1 addition, earlier than that found in OGM alone. Addition of a phospholipase C (PLC) inhibitor also prevented sFRP-stimulated increases in CXCL8 mRNA. siRNA technology targeting the Fzd-2 and 5 and the non-canonical Fzd co-receptor RoR2 also significantly decreased sFRP1/2-stimulated CXCL8 mRNA levels.
CONCLUSION: CXC chemokine expression in hMSCs is controlled in part by sFRPs signaling through non-canonical Wnt involving Fzd2/5 and the ERK and PLC pathways.
Collapse
|
181
|
Glycogen synthase kinase 3 inhibitor protects against microvascular hyperpermeability following hemorrhagic shock. J Trauma Acute Care Surg 2015; 79:609-16. [PMID: 26402535 DOI: 10.1097/ta.0000000000000807] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hemorrhagic shock (HS)-induced microvascular hyperpermeability involves disruption of endothelial cell adherens junctions leading to increase in paracellular permeability. β-Catenin, an integral component of the adherens junctional complex and Wnt pathway, and caspase 3 via its apoptotic signaling regulate endothelial cell barrier integrity. We have hypothesized that inhibiting phosphorylation of β-catenin and caspase 3 activity using glycogen synthase kinase 3-specific inhibitor SB216763 would attenuate microvascular hyperpermeability following HS. METHODS In Sprague-Dawley rats, HS was induced by withdrawing blood to reduce mean arterial pressure to 40 mm Hg for 60 minutes followed by resuscitation. Rats were given SB216763 (600 μg/kg) intravenously 10 minutes before shock. To study microvascular permeability, the rats were intravenously injected with fluorescein isothiocyanate (FITC)-albumin (50 mg/kg), and its flux across the mesenteric postcapillary venules was determined using intravital microscopy. In cell culture studies, rat lung microvascular endothelial cell monolayers grown on Transwell plates were pretreated with SB216763 (5 μM) followed by BAK (5 μg/mL) and caspase 3 (5 μg/mL) protein transfection. FITC-albumin (5 mg/mL) flux across cell monolayers indicates change in monolayer permeability. Activity of canonical Wnt pathway was determined by luciferase assay. Caspase 3 enzyme activity was assayed fluorometrically. RESULTS The HS group showed significant increase in FITC-albumin extravasation (p < 0.05) compared with sham. SB216763 significantly decrease HS-induced FITC-albumin extravasation (p < 0.05). Pretreatment with SB216763 protected against a BAK-induced increase in rat lung microvascular endothelial cell monolayer permeability and caspase 3 activity but failed to show similar results with a caspase 3-induced increase in monolayer permeability. Wnt3a treatment showed an increase in β-catenin-dependent T-cell factor-mediated transcription. CONCLUSION Inhibiting phosphorylation of β-catenin and caspase 3 activity using glycogen synthase kinase 3-specific inhibitor SB216763 help regulates HS-induced microvascular hyperpermeability.
Collapse
|
182
|
Felber K, Elks PM, Lecca M, Roehl HH. Expression of osterix Is Regulated by FGF and Wnt/β-Catenin Signalling during Osteoblast Differentiation. PLoS One 2015; 10:e0144982. [PMID: 26689368 PMCID: PMC4686927 DOI: 10.1371/journal.pone.0144982] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/26/2015] [Indexed: 01/24/2023] Open
Abstract
Osteoblast differentiation from mesenchymal cells is regulated by multiple signalling pathways. Here we have analysed the roles of Fibroblast Growth Factor (FGF) and canonical Wingless-type MMTV integration site (Wnt/β-Catenin) signalling pathways on zebrafish osteogenesis. We have used transgenic and chemical interference approaches to manipulate these pathways and have found that both pathways are required for osteoblast differentiation in vivo. Our analysis of bone markers suggests that these pathways act at the same stage of differentiation to initiate expression of the osteoblast master regulatory gene osterix (osx). We use two independent approaches that suggest that osx is a direct target of these pathways. Firstly, we manipulate signalling and show that osx gene expression responds with similar kinetics to that of known transcriptional targets of the FGF and Wnt pathways. Secondly, we have performed ChIP with transcription factors for both pathways and our data suggest that a genomic region in the first intron of osx mediates transcriptional activation. Based upon these data, we propose that FGF and Wnt/β-Catenin pathways act in part by directing transcription of osx to promote osteoblast differentiation at sites of bone formation.
Collapse
Affiliation(s)
- Katharina Felber
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Philip M. Elks
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Maria Lecca
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Henry H. Roehl
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
- * E-mail:
| |
Collapse
|
183
|
Bukowska J, Ziecik AJ, Laguna J, Gawronska-Kozak B, Bodek G. The Importance of the Canonical Wnt Signaling Pathway in the Porcine Endometrial Stromal Stem/Progenitor Cells: Implications for Regeneration. Stem Cells Dev 2015; 24:2873-85. [PMID: 26414529 PMCID: PMC4683563 DOI: 10.1089/scd.2015.0078] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022] Open
Abstract
The regenerative ability of the endometrium is strongly associated with the presence of adult stem/progenitor cells. Purposes of the present study were (1) to establish the presence of stem/progenitor cells in porcine endometrial stroma using a clonogenic assay and (2) to investigate whether the canonical Wnt pathway affects the potential of stem/progenitor cells to undergo self-renewal or differentiation. The utility of endometrial stromal clones as a model for stem/progenitor studies was evaluated based on these cells' increased expression of mesenchymal stem cell (MSC) marker genes, including CD29, CD73, CD90, and CD105, compared with primary cultured cells. Small molecules were introduced to activate (BIO) or inhibit (XAV939) the canonical Wnt pathway during stromal clone formation. Cloning efficiency assays revealed that activation of the Wnt/β-catenin pathway promoted formation of more differentiated small clones. Moreover, activation of the Wnt/β-catenin pathway decreased, whereas inhibition of the pathway increased MSC marker expression. Additionally, we confirmed the importance of canonical Wnt pathway stimulation in endometrial stromal cells through observing the appropriate changes in β-catenin cellular localization. These data indicate that modulation of the canonical Wnt pathway effects the process of regeneration in the porcine endometrium during the course of the estrous cycle.
Collapse
Affiliation(s)
- Joanna Bukowska
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Adam Janusz Ziecik
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Laguna
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Department of Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Gabriel Bodek
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
184
|
miR-140-5p regulates adipocyte differentiation by targeting transforming growth factor-β signaling. Sci Rep 2015; 5:18118. [PMID: 26657345 PMCID: PMC4676041 DOI: 10.1038/srep18118] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Recent emerging studies of miRNAs in adipocyte commitment provide new insights to understand the molecular basis of adipogenesis. The current study indicated that miR-140-5p was altered in primary cultured marrow stromal cells and established progenitor lines after adipogenic and/or osteogenic treatment. miR-140-5p was increased in adipose tissue in db/db obese mice vs. lean mice. Supplementing miR-140-5p activity induced stromal cell ST2 and preadipocyte 3T3-L1 to differentiate into mature adipocytes. Conversely, inhibition of the endogenous miR-140-5p repressed ST2 and 3T3-L1 to fully differentiate. By contrast, knockdown of the endogenous miR-140-5p enhanced osteoblast differentiation. Transforming growth factor-β receptor I (Tgfbr1) was shown to be a direct target of miR-140-5p. Supplementing miR-140-5p in ST2 reduced the level of TGFBR1 protein, while suppression of endogenous miR-140-5p increased TGFBR1. Overexpression of Tgfbr1 inhibited, whereas knockdown of Tgfbr1 promoted adipogenic differentiation of ST2 cells. Further investigation of mechanisms that control miR-140-5p expression revealed that C/EBPα induced transcriptional activity of the miR-140-5p promoter. Removal of the putative response element of C/EBP from the promoter abolished the enhancement of the promoter activity by C/EBPα, suggesting that C/EBPα transcriptionally controls miR-140-5p expression. Taken together, our study provides evidences that miR-140-5p regulates adipocyte differentiation through a C/EBP/miR-140-5p/TGFBR1 regulatory feedback loop.
Collapse
|
185
|
Yu F, Liu Z, Tong Z, Zhao Z, Liang H. Soybean isoflavone treatment induces osteoblast differentiation and proliferation by regulating analysis of Wnt/β-catenin pathway. Gene 2015; 573:273-7. [PMID: 26190158 DOI: 10.1016/j.gene.2015.07.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 06/24/2015] [Accepted: 07/15/2015] [Indexed: 10/23/2022]
Abstract
It has been reported that soybean isoflavones (SI) have anti-bone resorptive activity in vivo. However, little is known about the cellular and molecular mechanisms of this effect. In this study, we investigated the effects of SI on osteoblast differentiation and proliferation. Results showed that SI promoted osteoblast proliferation and differentiation which could be marked as the formation of hydroxyapatite crystals or extracellular matrix and increased ALP level. SI treatment markedly decreased RANKL levels and increased OPG levels in osteoblast cells. SI resulted in activation of the Wnt/β-catenin pathway. These results demonstrate that SI may be a useful remedy for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Fang Yu
- The Second Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Zhonghua Liu
- Orthopedic Department, Changchun University of Traditional Chinese Medicine Affiliated Hospital, Changchun 130021, People's Republic of China
| | - Zhihong Tong
- Hands and Feet Microsurgery, The Second Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Zhengnan Zhao
- Hands and Feet Microsurgery, The Second Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Haidong Liang
- Hands and Feet Microsurgery, The Second Hospital of Dalian Medical University, Dalian 116023, People's Republic of China.
| |
Collapse
|
186
|
Janeczek AA, Tare RS, Scarpa E, Moreno-Jimenez I, Rowland CA, Jenner D, Newman TA, Oreffo ROC, Evans ND. Transient Canonical Wnt Stimulation Enriches Human Bone Marrow Mononuclear Cell Isolates for Osteoprogenitors. Stem Cells 2015; 34:418-30. [PMID: 26573091 PMCID: PMC4981914 DOI: 10.1002/stem.2241] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/01/2015] [Indexed: 12/27/2022]
Abstract
Activation of the canonical Wnt signaling pathway is an attractive anabolic therapeutic strategy for bone. Emerging data suggest that activation of the Wnt signaling pathway promotes bone mineral accrual in osteoporotic patients. The effect of Wnt stimulation in fracture healing is less clear as Wnt signaling has both stimulatory and inhibitory effects on osteogenesis. Here, we tested the hypothesis that transient Wnt stimulation promotes the expansion and osteogenesis of a Wnt‐responsive stem cell population present in human bone marrow. Bone marrow mononuclear cells (BMMNCs) were isolated from patients undergoing hip arthroplasty and exposed to Wnt3A protein. The effect of Wnt pathway stimulation was determined by measuring the frequency of stem cells within the BMMNC populations by fluorescence‐activated cell sorting and colony forming unit fibroblast (CFU‐F) assays, before determining their osteogenic capacity in in vitro differentiation experiments. We found that putative skeletal stem cells in BMMNC isolates exhibited elevated Wnt pathway activity compared with the population as whole. Wnt stimulation resulted in an increase in the frequency of skeletal stem cells marked by the STRO‐1bright/Glycophorin A− phenotype. Osteogenesis was elevated in stromal cell populations arising from BMMNCs transiently stimulated by Wnt3A protein, but sustained stimulation inhibited osteogenesis in a concentration‐dependent manner. These results demonstrate that Wnt stimulation could be used as a therapeutic approach by transient targeting of stem cell populations during early fracture healing, but that inappropriate stimulation may prevent osteogenesis. Stem Cells2016;34:418–430
Collapse
Affiliation(s)
- Agnieszka A Janeczek
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Rahul S Tare
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Edoardo Scarpa
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Ines Moreno-Jimenez
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Caroline A Rowland
- Microbiology group, Chemical, Biological and Radiological Division, Salisbury, United Kingdom
| | - Dominic Jenner
- Microbiology group, Chemical, Biological and Radiological Division, Salisbury, United Kingdom
| | - Tracey A Newman
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Nicholas D Evans
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
187
|
Mahmood S, Bhatti A, Syed NA, John P. The microRNA regulatory network: a far-reaching approach to the regulate the Wnt signaling pathway in number of diseases. J Recept Signal Transduct Res 2015; 36:310-8. [PMID: 26523375 DOI: 10.3109/10799893.2015.1080273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Wnt signaling pathway plays an important role in cell renewal, tumorigenesis, organogenesis, bone formation and bone resorption. Wnt signaling pathway is divided into two outlets: Wnt-β-catenin pathway (canonical pathway) and Wnt-calcium pathway (non-canonical pathway). miRNAs play a key role in the regulation of Wnt signaling pathway. In this review, we highlight the basic indulgent of miRNAs-mediated regulation of Wnt signaling pathway. We focus on the role of miRNAs at different levels of Wnt signaling: signaling molecules, their associated signaling proteins, regulatory proteins, transcription factors and related cytokines. Finally, we concluded that these multiple levels of targeting may have diagnostic potential as well as therapeutic prospective in future treatment.
Collapse
Affiliation(s)
- Shahid Mahmood
- a Immunogenetic Lab, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Science and Technology (NUST) , Islamabad , Pakistan
| | - Attya Bhatti
- a Immunogenetic Lab, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Science and Technology (NUST) , Islamabad , Pakistan
| | - Nida Ali Syed
- a Immunogenetic Lab, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Science and Technology (NUST) , Islamabad , Pakistan
| | - Peter John
- a Immunogenetic Lab, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Science and Technology (NUST) , Islamabad , Pakistan
| |
Collapse
|
188
|
Barter MJ, Tselepi M, Gómez R, Woods S, Hui W, Smith GR, Shanley DP, Clark IM, Young DA. Genome-Wide MicroRNA and Gene Analysis of Mesenchymal Stem Cell Chondrogenesis Identifies an Essential Role and Multiple Targets for miR-140-5p. Stem Cells 2015; 33:3266-80. [PMID: 26175215 PMCID: PMC4737122 DOI: 10.1002/stem.2093] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/20/2015] [Accepted: 06/01/2015] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are abundantly expressed in development where they are critical determinants of cell differentiation and phenotype. Accordingly miRNAs are essential for normal skeletal development and chondrogenesis in particular. However, the question of which miRNAs are specific to the chondrocyte phenotype has not been fully addressed. Using microarray analysis of miRNA expression during mesenchymal stem cell chondrogenic differentiation and detailed examination of the role of essential differentiation factors, such as SOX9, TGF-β, and the cell condensation phase, we characterize the repertoire of specific miRNAs involved in chondrocyte development, highlighting in particular miR-140 and miR-455. Further with the use of mRNA microarray data we integrate miRNA expression and mRNA expression during chondrogenesis to underline the particular importance of miR-140, especially the -5p strand. We provide a detailed identification and validation of direct targets of miR-140-5p in both chondrogenesis and adult chondrocytes with the use of microarray and 3'UTR analysis. This emphasizes the diverse array of targets and pathways regulated by miR-140-5p. We are also able to confirm previous experimentally identified targets but, additionally, identify a novel positive regulation of the Wnt signaling pathway by miR-140-5p. Wnt signaling has a complex role in chondrogenesis and skeletal development and these findings illustrate a previously unidentified role for miR-140-5p in regulation of Wnt signaling in these processes. Together these developments further highlight the role of miRNAs during chondrogenesis to improve our understanding of chondrocyte development and guide cartilage tissue engineering.
Collapse
Affiliation(s)
- Matt J. Barter
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Maria Tselepi
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Rodolfo Gómez
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Steven Woods
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Wang Hui
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Graham R. Smith
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Daryl P. Shanley
- Institute for Ageing and HealthNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Ian M. Clark
- School of Biological SciencesUniversity of East AngliaNorwichUnited Kingdom
| | - David A. Young
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| |
Collapse
|
189
|
Ahmadzadeh A, Norozi F, Shahrabi S, Shahjahani M, Saki N. Wnt/β-catenin signaling in bone marrow niche. Cell Tissue Res 2015; 363:321-35. [PMID: 26475718 DOI: 10.1007/s00441-015-2300-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/20/2015] [Indexed: 12/14/2022]
Abstract
The bone marrow (BM) niche is a specific physiological environment for hematopoietic and non-hematopoietic stem cells (HSCs). Several signaling pathways (including Wnt/β-catenin) regulate various aspects of stem cell growth, function and death in the BM niche. In addition, the canonical Wnt pathway is crucial for directing self-renewal and differentiation as important mechanisms in many types of stem cells. We review the role of the Wnt/β-catenin pathway in the BM niche and its importance in stem cells. Relevant literature was identified by a PubMed search (1997-2014) of English-language literature by using the following keywords: BM niche, Wnt/β-catenin signaling, osteoblast, osteoclast and bone disease. The Wnt/β-catenin pathway regulates the stability of the β-catenin proto-oncogene. The stabilized β-catenin then translocates to the nucleus, forming a β-catenin-TCF/LEF complex regulating the transcription of specific target genes. Stem cells require β-catenin to mediate their response to Wnt signaling for maintenance and transition from the pluripotent state during embryogenesis. In adult stem cells, Wnt signaling functions at various hierarchical levels to contribute to the specification of the diverse tissues. Aberrant Wnt/β-catenin signaling and its downstream transcriptional regulators are observed in several malignant stem cells and human cancers. Because Wnt signaling can maintain stem cells and cancer cells, the ability to modulate the Wnt pathway either positively or negatively may be of therapeutic relevance. The controlled activation of Wnt signaling might allow us to enhance stem and progenitor cell activity when regeneration is needed.
Collapse
Affiliation(s)
- Ahmad Ahmadzadeh
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Norozi
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Shahjahani
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
190
|
Tomé M, Sepúlveda JC, Delgado M, Andrades JA, Campisi J, González MA, Bernad A. miR-335 correlates with senescence/aging in human mesenchymal stem cells and inhibits their therapeutic actions through inhibition of AP-1 activity. Stem Cells 2015; 32:2229-44. [PMID: 24648336 DOI: 10.1002/stem.1699] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 02/25/2014] [Accepted: 03/02/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs, small noncoding RNAs, regulate gene expression primarily at the posttranscriptional level. We previously found that miR-335 is critically involved in the regulation and differentiation capacity of human mesenchymal stem cells (hMSCs) in vitro. In this study, we investigated the significance of miR-335 for the therapeutic potential of hMSCs. Analysis of hMSCs in ex vivo culture demonstrated a significant and progressive increase in miR-335 that is prevented by telomerase. Expression levels of miR-335 were also positively correlated with donor age of hMSCs, and were increased by stimuli that induce cell senescence, such as γ-irradiation and standard O2 concentration. Forced expression of miR-335 resulted in early senescence-like alterations in hMSCs, including: increased SA-β-gal activity and cell size, reduced cell proliferation capacity, augmented levels of p16 protein, and the development of a senescence-associated secretory phenotype. Furthermore, overexpression of miR-335 abolished the in vivo chondro-osseous potential of hMSCs, and disabled their immunomodulatory capacity in a murine experimental model of lethal endotoxemia. These effects were accompanied by a severely reduced capacity for cell migration in response to proinflammatory signals and a marked reduction in Protein Kinase D1 phosphorylation, resulting in a pronounced decrease of AP-1 activity. Our results demonstrate that miR-335 plays a key role in the regulation of reparative activities of hMSCs and suggests that it might be considered a marker for the therapeutic potency of these cells in clinical applications.
Collapse
Affiliation(s)
- María Tomé
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
191
|
Lu J, Duan Y, Zhang M, Wu M, Wang Y. Expression of Wnt3a, Wnt10b, β-catenin and DKK1 in periodontium during orthodontic tooth movement in rats. Acta Odontol Scand 2015; 74:217-23. [PMID: 26414930 DOI: 10.3109/00016357.2015.1090011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To investigate the expression of Wnt3a, Wnt10b, β-catenin and DKK1 in the periodontal ligament (PDL) during orthodontic tooth movement (OTM) in rats. MATERIALS AND METHODS Nickel-titanium closed-coil springs were used to deliver an initial 50 g mesial force to the left maxillary first molars in 30 rats. The force was kept constant for 1, 3, 5, 7, 10 and 14 days until the animals were sacrificed. The right maxillary molars without force application served as control. Paraffin-embedded sections of the upper jaws were prepared for histological and immunohistochemical analyses to detect Wnt3a, Wnt10b, β-catenin and DKK1 expression in PDL. RESULTS Wnt3a, Wnt10b, β-catenin and DKK1 were expressed on both the ipsilateral and contralateral sides of PDL in each group. After the application of orthodontic force, the expression of β-catenin and DKK1 was initially increased and then decreased on both sides, with maximal levels of expression at day 7 and day 10, respectively. On the compression side, Wnt3a and Wnt10b levels started to increase at day 5, while on the tension side, these two molecules began to increase at day 1. Furthermore, the expression levels of Wnt3a, Wnt10b, and β-catenin were much stronger on the tension side than on the compression side at any of the observation points, while DKK1 level was much higher on the compression side. CONCLUSION Wnt3a, Wnt10b, β-catenin and DKK1 expression may be related to the periodontal tissue remodeling following the application of an orthodontic force in rats. These observations suggest that the Wnt/β-catenin signaling pathway may play a crucial role in periodontal tissue remodeling during OTM.
Collapse
Affiliation(s)
- Juan Lu
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Yingying Duan
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Miaomiao Zhang
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Mingming Wu
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Ying Wang
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| |
Collapse
|
192
|
Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, Pervaiz S, Dharmarajan A. Multi-lineage differentiation of mesenchymal stem cells - To Wnt, or not Wnt. Int J Biochem Cell Biol 2015; 68:139-47. [PMID: 26410622 DOI: 10.1016/j.biocel.2015.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent precursor cells originating from several adult connective tissues. MSCs possess the ability to self-renew and differentiate into several lineages, and are recognized by the expression of unique cell surface markers. Several lines of evidence suggest that various signal transduction pathways and their interplay regulate MSC differentiation. To that end, a critical player in regulating MSC differentiation is a group of proteins encoded by the Wnt gene family, which was previously known for influencing various stages of embryonic development and cell fate determination. As MSCs have gained significant clinical attention for their potential applications in regenerative medicine, it is imperative to unravel the mechanisms by which molecular regulators control differentiation of MSCs for designing cell-based therapeutics. It is rather coincidental that the functional outcome(s) of Wnt-induced signals share similarities with cellular redox-mediated networks from the standpoint of MSC biology. Furthermore, there is evidence for a crosstalk between Wnt and redox signalling, which begs the question whether Wnt-mediated differentiation signals involve the intermediary role of reactive oxygen species. In this review, we summarize the impact of Wnt signalling on multi-lineage differentiation of MSCs, and attempt to unravel the intricate interplay between Wnt and redox signals.
Collapse
Affiliation(s)
- Malini Visweswaran
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Sebastian Pohl
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Rodney Dilley
- Ear Sciences Centre, University of Western Australia and Ear Science Institute Australia, Perth, Western Australia 6008, Australia
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
193
|
Katagiri W, Osugi M, Kawai T, Hibi H. Secreted Frizzled-Related Protein Promotes Bone Regeneration by Human Bone Marrow-Derived Mesenchymal Stem Cells. Int J Mol Sci 2015; 16:23250-8. [PMID: 26404242 PMCID: PMC4632696 DOI: 10.3390/ijms161023250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/10/2015] [Accepted: 09/23/2015] [Indexed: 11/16/2022] Open
Abstract
Secreted frizzled-related protein (sFRP)-3 is a negative regulator of Wnt signaling in human mesenchymal stem cells (hMSCs). The present study investigated the effects sFRP-3 on osteogenic differentiation by assessing osteogenic gene expression in hMSCs in vitro and by examining bone regeneration in a rat bone defect model. sFRP-3 treatment induced osteogenic differentiation in hMSCs as determined by alkaline phosphatase, collagen type I, osteocalcin, and Runt-related transcription factor 2 gene expression. hMSCs with or without sFRP-3 were implanted into a rat calvarial bone defect; a radiographic analysis by micro-computed tomography and histological analysis 4 and 8 weeks after implantation showed greater bone regeneration in the sFRP(+) than in the sFRP(−) group. These results suggest that modulation of Wnt signaling contributes to osteogenic differentiation in hMSCs. Specifically, sFRP-3 induces osteoblastic differentiation of cultured MSCs and bone regeneration in a calvarial bone defect, suggesting that it can be a useful agent for the treatment of bone defects.
Collapse
Affiliation(s)
- Wataru Katagiri
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Masashi Osugi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Takamasa Kawai
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| |
Collapse
|
194
|
DKK1 rescues osteogenic differentiation of mesenchymal stem cells isolated from periodontal ligaments of patients with diabetes mellitus induced periodontitis. Sci Rep 2015; 5:13142. [PMID: 26278788 PMCID: PMC4538385 DOI: 10.1038/srep13142] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/21/2015] [Indexed: 12/11/2022] Open
Abstract
Multiple studies have shown that diabetes mellitus is an established risk factor for periodontitis. Recently mesenchymal stem cells derived from periodontal ligament (PDLSCs) have been utilized to reconstruct tissues destroyed by chronic inflammation. However, impact of periodontitis with diabetes mellitus on PDLSCs and mechanisms mediating effects of complex microenvironments remain poorly understood. In this study, we found multiple differentiation potential of PDLSCs from chronic periodontitis with diabetes mellitus donors (D-PDLSCs) was damaged significantly. Inhibition of NF-κB signaling could rescue osteogenic potential of PDLSCs from simple chronic periodontitis patients (P-PDLSCs), whereas did not promote D-PDLSCs osteogenesis. In addition, we found expression of DKK1 in D-PDLSCs did not respond to osteogenic signal and decreased osteogenic potential of D-PDLSCs treated with DKK1 could be reversed. To further elucidate different character between P-PDLSCs and D-PDLSCs, we treated PDLSCs with TNF-α and advanced glycation end products (AGEs), and find out AGEs which enhance effect of TNF-α in PDLSCs might mediate special personality of D-PDLSCs. The adverse effect of AGEs in PDLSCs could be reversed when PDLSCs were treated with DKK1. These results suggested DKK1 mediating WNT signaling might be a therapy target to rescue potential of PDLSCs in periodontitis with diabetes mellitus.
Collapse
|
195
|
Díaz-Rodríguez P, Gómez-Amoza JL, Landin M. The synergistic effect of VEGF and biomorphic silicon carbides topography on
in vivo
angiogenesis and human bone marrow derived mesenchymal stem cell differentiation. Biomed Mater 2015; 10:045017. [DOI: 10.1088/1748-6041/10/4/045017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
196
|
Developmental Stage-Dependent Effects of Leukemia Inhibitory Factor on Adipocyte Differentiation of Murine Bone Marrow Stromal Cells. Cell Biochem Biophys 2015; 74:11-7. [DOI: 10.1007/s12013-015-0703-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
197
|
Li Y, Liu F, Zhang Z, Zhang M, Cao S, Li Y, Zhang L, Huang X, Xu Y. Bone marrow mesenchymal stem cells could acquire the phenotypes of epithelial cells and accelerate vaginal reconstruction combined with small intestinal submucosa. Cell Biol Int 2015; 39:1225-33. [PMID: 26018040 DOI: 10.1002/cbin.10495] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/16/2015] [Indexed: 11/12/2022]
Abstract
Grafting material for vaginal reconstruction commonly includes the bowel, peritoneum, skin, and amniotic membrane. Bone marrow mesenchymal stem cells (MSCs) have the potential of multilineage differentiation into a variety of cells and have been widely explored in tissue engineering. In the current study, we examined whether MSCs could be differentiated to vaginal epithelial cells (VECs) upon co-culturing with VECs. We also examined whether Wnt/β-catenin signaling pathway is implicated in such differentiation. Co-culture of MSCs with VECs using a transwell insert system (with no direct contact) induced the expression of VECs marker AE1/AE3 in MSCs. MSCs combined with small intestinal submucosa (SIS) scaffold were implanted in place of the native vagina in rats to observe the implications for vaginal reconstruction in vivo. Anatomic repair of neovagina was assessed by histological staining for H/E and Masson's Trichrome. GSK-3β and β-catenin, main members of Wnt/β-catenin signaling pathway, in MSCs were increased upon co-culturing with VECs. Exposure of co-cultured MSCs to a Wnt/β-catenin signaling activator, lithium chloride (LiCl, 20 µM) increased phosphorylated GSK-3β and β-catenin and enhanced expression of AE1/AE3. In vivo-grafted cells displayed significant matrix infiltration and expressed epithelial markers in neovagina. These findings suggest that MSCs could acquire the phenotype of VECs when co-cultured with VECs, possibly via activation of Wnt/β-catenin signaling. MSCs provide an alternative cell source for potential use in vaginal tissue engineering.
Collapse
Affiliation(s)
- Yanan Li
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Fangfang Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Zhiqiang Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Mingle Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Shanjin Cao
- Mount Vernon Hospital Affiliated With New York Medical College, 12 N 7th Avenue, Mount Vernon, New York, 10550
| | - Yachai Li
- Department of Obstetrics and Gynecology, Hospital of Hebei University, 213 Yuhua East Road, Baoding, 071000, China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050000, China
| |
Collapse
|
198
|
Zhang Y, Li J, Davis ME, Pei M. Delineation of in vitro chondrogenesis of human synovial stem cells following preconditioning using decellularized matrix. Acta Biomater 2015; 20:39-50. [PMID: 25861949 DOI: 10.1016/j.actbio.2015.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/17/2015] [Accepted: 04/02/2015] [Indexed: 11/27/2022]
Abstract
As a tissue-specific stem cell for chondrogenesis, synovium-derived stem cells (SDSCs) are a promising cell source for cartilage repair. However, a small biopsy can only provide a limited number of cells. Cell senescence from both in vitro expansion and donor age presents a big challenge for stem cell based cartilage regeneration. Here we found that expansion on decellularized extracellular matrix (dECM) full of three-dimensional nanostructured fibers provided SDSCs with unique surface profiles, low elasticity but large volume as well as a fibroblast-like shape. dECM expanded SDSCs yielded larger pellets with intensive staining of type II collagen and sulfated glycosaminoglycans compared to those grown on plastic flasks while SDSCs grown in ECM yielded 28-day pellets with minimal matrix as evidenced by pellet size and chondrogenic marker staining, which was confirmed by both biochemical data and real-time PCR data. Our results also found lower levels of inflammatory genes in dECM expanded SDSCs that might be responsible for enhanced chondrogenic differentiation. Despite an increase in type X collagen in chondrogenically induced cells, dECM expanded cells had significantly lower potential for endochondral bone formation. Wnt and MAPK signals were actively involved in both expansion and chondrogenic induction of dECM expanded cells. Since young and healthy people can be potential donors for this matrix expansion system and decellularization can minimize immune concerns, human SDSCs expanded on this future commercially available dECM could be a potential cell source for autologous cartilage repair.
Collapse
Affiliation(s)
- Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
| | - Mary E Davis
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
199
|
Park YB, Seo S, Kim JA, Heo JC, Lim YC, Ha CW. Effect of chondrocyte-derived early extracellular matrix on chondrogenesis of placenta-derived mesenchymal stem cells. ACTA ACUST UNITED AC 2015; 10:035014. [PMID: 26107298 DOI: 10.1088/1748-6041/10/3/035014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The extracellular matrix (ECM) surrounding cells contains a variety of proteins that provide structural support and regulate cellular functions. Previous studies have shown that decellularized ECM isolated from tissues or cultured cells can be used to improve cell differentiation in tissue engineering applications. In this study we evaluated the effect of decellularized chondrocyte-derived ECM (CDECM) on the chondrogenesis of human placenta-derived mesenchymal stem cells (hPDMSCs) in a pellet culture system. After incubation with or without chondrocyte-derived ECM in chondrogenic medium for 1 or 3 weeks, the sizes and wet masses of the cell pellets were compared with untreated controls (hPDMSCs incubated in chondrogenic medium without chondrocyte-derived ECM). In addition, histologic analysis of the cell pellets (Safranin O and collagen type II staining) and quantitative reverse transcription-PCR analysis of chondrogenic markers (aggrecan, collagen type II, and SOX9) were carried out. Our results showed that the sizes and masses of hPDMSC pellets incubated with chondrocyte-derived ECM were significantly higher than those of untreated controls. Differentiation of hPDMSCs (both with and without chondrocyte-derived ECM) was confirmed by Safranin O and collagen type II staining. Chondrogenic marker expression and glycosaminoglycan (GAG) levels were significantly higher in hPDMSC pellets incubated with chondrocyte-derived ECM compared with untreated controls, especially in cells precultured with chondrocyte-derived ECM for 7 d. Taken together, these results demonstrate that chondrocyte-derived ECM enhances the chondrogenesis of hPDMSCs, and this effect is further increased by preculture with chondrocyte-derived ECM. This preculture method for hPDMSC chondrogenesis represents a promising approach for cartilage tissue engineering.
Collapse
|
200
|
Nemoto E, Sakisaka Y, Tsuchiya M, Tamura M, Nakamura T, Kanaya S, Shimonishi M, Shimauchi H. Wnt3a signaling induces murine dental follicle cells to differentiate into cementoblastic/osteoblastic cells via an osterix-dependent pathway. J Periodontal Res 2015; 51:164-74. [PMID: 26095156 DOI: 10.1111/jre.12294] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Dental follicle cells, putative progenitor cells for cementoblasts, osteoblasts and periodontal ligament cells, interplay with Hertwig's epithelial root sheath (HERS) cells during tooth root formation, in which HERS is considered to have an inductive role in initiating cementogenesis by epithelial-mesenchymal interaction. However, the specific mechanisms controlling the cementoblast/osteoblast differentiation of dental follicle cells are not fully understood. Canonical Wnt signaling has been implicated in increased bone formation by controlling mesenchymal stem cell or osteoblastic cell functions. This study examined the possible expression of canonical Wnt ligand in HERS and the role of Wnt signaling during the cementoblast/osteoblast differentiation of dental follicle cells. MATERIAL AND METHODS The expression of Wnt3a, a representative canonical Wnt ligand, in HERS was assessed by immunohistochemistry. The differentiation and function of immortalized murine dental follicle cells were evaluated by measuring alkaline phosphatase (ALP, Alpl) activity and osteogenic gene expression. RESULTS We identified the expression of Wnt3a in HERS during mouse tooth root development by immunohistochemistry as well as in cultured human epithelial rest cells of Malassez by real-time polymerase chain reaction, while no expression of Wnt3a was detected in cultured dental mesenchymal cells. Exposure of immortalized murine dental follicle cells to Wnt3a-induced ALP activity as well as expression of the Alpl gene. Pretreatment of cells with Dickkopf-1, a potent canonical Wnt antagonist, markedly attenuated the effect of Wnt3a on ALP expression. Furthermore, Wnt3a induced transcriptional activity of runt-related transcription factor 2 (Runx2) and expression of osterix at gene and/or protein levels. Treatment with osterix-small interfering RNA significantly inhibited Wnt3a-induced ALP expression at gene and protein levels. CONCLUSION These findings suggest that HERS has a potential role in stimulating cementoblast/osteoblast differentiation of dental follicle cells via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- E Nemoto
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Y Sakisaka
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - M Tsuchiya
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Tohoku Fukushi University, Sendai, Japan
| | - M Tamura
- Department of Biochemistry and Molecular Biology, Hokkaido University Graduate School of Dentistry, Sapporo, Japan
| | - T Nakamura
- Department of Pediatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - S Kanaya
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - M Shimonishi
- Department of Comprehensive Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - H Shimauchi
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|