1
|
Mei R, Wan Z, Yang C, Shen X, Wang R, Zhang H, Yang R, Li J, Song Y, Su H. Advances and clinical challenges of mesenchymal stem cell therapy. Front Immunol 2024; 15:1421854. [PMID: 39100671 PMCID: PMC11294097 DOI: 10.3389/fimmu.2024.1421854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
In recent years, cell therapy has provided desirable properties for promising new drugs. Mesenchymal stem cells are promising candidates for developing genetic engineering and drug delivery strategies due to their inherent properties, including immune regulation, homing ability and tumor tropism. The therapeutic potential of mesenchymal stem cells is being investigated for cancer therapy, inflammatory and fibrotic diseases, among others. Mesenchymal stem cells are attractive cellular carriers for synthetic nanoparticles for drug delivery due to their inherent homing ability. In this review, we comprehensively discuss the various genetic and non-genetic strategies of mesenchymal stem cells and their derivatives in drug delivery, tumor therapy, immune regulation, tissue regeneration and other fields. In addition, we discuss the current limitations of stem cell therapy and the challenges in clinical translation, aiming to identify important development areas and potential future directions.
Collapse
Affiliation(s)
- Ruiyan Mei
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Zhuo Wan
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Cheng Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xiangjing Shen
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haihua Zhang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Rui Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
2
|
Iwobi N, Sparks NR. Endocrine Disruptor-Induced Bone Damage Due to Hormone Dysregulation: A Review. Int J Mol Sci 2023; 24:ijms24098263. [PMID: 37175969 PMCID: PMC10179611 DOI: 10.3390/ijms24098263] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Hormones are indispensable for bone development, growth, and maintenance. While many of the genes associated with osteogenesis are well established, it is the recent findings in endocrinology that are advancing the fields of bone biology and toxicology. Endocrine-disrupting chemicals (EDCs) are defined as chemicals that interfere with the function of the endocrine system. Here, we report recent discoveries describing key hormone pathways involved in osteogenesis and the EDCs that alter these pathways. EDCs can lead to bone morphological changes via altering hormone receptors, signaling pathways, and gene expression. The objective of this review is to highlight the recent discoveries of the harmful effects of environmental toxicants on bone formation and the pathways impacted. Understanding the mechanisms of how EDCs interfere with bone formation contributes to providing a comprehensive toxicological profile of a chemical.
Collapse
Affiliation(s)
- Nneamaka Iwobi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California, Irvine, CA 92697, USA
| | - Nicole R Sparks
- Department of Occupational and Environmental Health, University of California, Irvine, CA 92697, USA
| |
Collapse
|
3
|
Wang D, Qi Y, Wang Z, Guo A, Xu Y, Zhang Y. Recent Advances in Animal Models, Diagnosis, and Treatment of Temporomandibular Joint Osteoarthritis. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:62-77. [PMID: 35994388 DOI: 10.1089/ten.teb.2022.0065] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a gradual degenerative jaw joint condition. Until recent years, TMJOA is still relatively unrecognized and ineffective to be treated. Appropriate animal models with reliable detection methods can help researchers understand the pathophysiology of TMJOA and find therapeutic options. In this study, we summarized common animal models of TMJOA created by chemical, surgical, mechanical, and genetical approaches. The relevant pathological symptoms and induction mechanisms were outlined. In addition, different pathological indicators, furthermore, emerging therapeutic regimens, such as intra-articular drug delivery and tissue engineering-based approaches to treat TMJOA based on these animal models, were summarized and updated. Understanding the physiology and pathogenesis of the TMJOA, together using various ways to diagnose the TMJOA, were elaborated, including imaging techniques, molecular techniques for detecting inflammatory cytokines, histochemical staining, and histomorphometry measures. A more reliable diagnosis will enable the development of new prevention and more effective treatment strategies and thereby improve the quality of life of TMJOA patients. Impact statement Temporomandibular joint osteoarthritis (TMJOA) affects 8 to 16 percent of the population worldwide. However, TMJOA is still relatively unrecognized and ineffective to be treated in the clinic. Appropriate animal models with reliable diagnostic methods can help researchers understand the pathophysiology of TMJOA and find therapeutic options. We herein summarized common animal models of TMJOA and various ways to diagnose the TMJOA. More importantly, emerging therapeutic regimens to treat TMJOA based on these animal models were summarized. With the aid of strategies listed, more effective treatment strategies will be developed and thereby improve the life quality of TMJOA patients.
Collapse
Affiliation(s)
- Dongyun Wang
- Stomatological Center of Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, China
| | - Yajie Qi
- Stomatological Center of Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, China.,Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
| | - Zhubing Wang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, China
| | - Anyun Guo
- Department of Joint Surgery, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Yingxin Xu
- Stomatological Center of Peking University Shenzhen Hospital, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, China
| | - Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Uniparental parthenogenetic embryonic stem cell derivatives adaptable for bone and cartilage regeneration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119379. [PMID: 36228838 DOI: 10.1016/j.bbamcr.2022.119379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
Cells with the desired phenotype and number are critical for regenerative medicine and tissue engineering. Uniparental parthenogenetic embryonic stem cells (pESCs) share fundamental properties with embryonic stem cells. This study aims to determine the viability of pESC-based tissue engineering for bone and cartilage reconstruction. The mouse pESCs were cultured in suspension to form embryoid bodies. An adherent cultivation approach was employed to obtain parthenogenetic embryonic mesenchymal stem cells (pMSCs) from the embryoid bodies. Then, the pMSCs were cultured in conditional media to differentiate into osteogenic and chondrogenic lineages. The pESC-derived osteoblasts and chondroblasts were seeded into coral and sodium alginate scaffolds, respectively. The cell-seeded scaffolds were implanted into dorsal subcutaneous pockets of nude mice to evaluate ectopic reconstruction of bone and cartilage. We demonstrated that pESCs display the capacity to differentiate into all three germ layers. The generated pMSCs were able to differentiate into osteogenic and chondrogenic lineages, which survived well after seeding into coral and alginate acid scaffolds. Six weeks after cell-scaffold implantation, gross inspection and histological examination revealed that ectopic bone and cartilage tissues had successfully regenerated in the specimen. According to the findings of this study, pESC derivatives have a high potential for bone and cartilage regeneration.
Collapse
|
5
|
Dienelt A, Keller KC, zur Nieden NI. High glucose impairs osteogenic differentiation of embryonic stem cells via early diversion of beta-catenin from Forkhead box O to T cell factor interaction. Birth Defects Res 2022; 114:1056-1074. [PMID: 36164276 PMCID: PMC9708100 DOI: 10.1002/bdr2.2085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Diabetes, which is characterized by an increase in blood glucose concentration, is accompanied by low bone turnover, increased fracture risk, and the formation of embryonic skeletal malformations. Yet, there are few studies elucidating the underlying alterations in signaling pathways leading to these osteogenic defects. We hypothesized here that bone formation deficiencies in a high glucose environment result from altered activity of beta-catenin (CTNNB1), a key contributor to osteogenic differentiation, dysregulation of which has also been implicated in the development of diabetes. METHODS To test this hypothesis, we used a previously established embryonic stem cell (ESC) model of differentiation that mimics the diabetic environment of the developing embryo. We differentiated murine ESCs within osteogenic-inducing media containing either high (diabetic) or low (physiological) levels of D-glucose and performed time course analyses to study the influence of high glucose on early and late bone cell differentiation. RESULTS Endpoint measures for osteogenic differentiation were reduced in a glucose-dependent manner and expression of precursor-specific markers altered at multiple time points. Furthermore, transcriptional activity of the lymphoid enhancer factor (LEF)/T cell factor (TCF) transcription factors during precursor formation stages was significantly elevated while levels of CTNNB1 complexed with Forkhead box O 3a (FOXO3a) declined. Modulation of AKT, a known upstream regulator of both LEF/TCF and FOXO3a, as well as CTNNB1 rescued some of the reductions in osteogenic output seen in the high glucose condition. CONCLUSIONS Within our in vitro model, we found a clear involvement of LEF/TCF and FOXO3a signaling pathways in the regulation of osteogenic differentiation, which may account for the skeletal deficiencies found in newborns of diabetic mothers.
Collapse
Affiliation(s)
- Anke Dienelt
- Department of Cell Therapy, Applied Stem Cell Technologies Unit, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Kevin C. Keller
- Department of Molecular, Cell and Systems Biology & Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| | - Nicole I. zur Nieden
- Department of Cell Therapy, Applied Stem Cell Technologies Unit, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Department of Molecular, Cell and Systems Biology & Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
6
|
Cun X, Hosta-Rigau L. Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2070. [PMID: 33092104 PMCID: PMC7590059 DOI: 10.3390/nano10102070] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising strategy to treat tissue and organ loss or damage caused by injury or disease. During the past two decades, mesenchymal stem cells (MSCs) have attracted a tremendous amount of interest in tissue engineering due to their multipotency and self-renewal ability. MSCs are also the most multipotent stem cells in the human adult body. However, the application of MSCs in tissue engineering is relatively limited because it is difficult to guide their differentiation toward a specific cell lineage by using traditional biochemical factors. Besides biochemical factors, the differentiation of MSCs also influenced by biophysical cues. To this end, much effort has been devoted to directing the cell lineage decisions of MSCs through adjusting the biophysical properties of biomaterials. The surface topography of the biomaterial-based scaffold can modulate the proliferation and differentiation of MSCs. Presently, the development of micro- and nano-fabrication techniques has made it possible to control the surface topography of the scaffold precisely. In this review, we highlight and discuss how the main topographical features (i.e., roughness, patterns, and porosity) are an efficient approach to control the fate of MSCs and the application of topography in tissue engineering.
Collapse
Affiliation(s)
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Nils Koppels Allé, Building 423, 2800 Kgs. Lyngby, Denmark;
| |
Collapse
|
7
|
Senile Osteoporosis: The Involvement of Differentiation and Senescence of Bone Marrow Stromal Cells. Int J Mol Sci 2020; 21:ijms21010349. [PMID: 31948061 PMCID: PMC6981793 DOI: 10.3390/ijms21010349] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
Senile osteoporosis has become a worldwide bone disease with the aging of the world population. It increases the risk of bone fracture and seriously affects human health. Unlike postmenopausal osteoporosis which is linked to menopause in women, senile osteoporosis is due to aging, hence, affecting both men and women. It is commonly found in people with more than their 70s. Evidence has shown that with age increase, bone marrow stromal cells (BMSCs) differentiate into more adipocytes rather than osteoblasts and undergo senescence, which leads to decreased bone formation and contributes to senile osteoporosis. Therefore, it is necessary to uncover the molecular mechanisms underlying the functional changes of BMSCs. It will benefit not only for understanding the senile osteoporosis development, but also for finding new therapies to treat senile osteoporosis. Here, we review the recent advances of the functional alterations of BMSCs and the related mechanisms during senile osteoporosis development. Moreover, the treatment of senile osteoporosis by aiming at BMSCs is introduced.
Collapse
|
8
|
Zhang K, Yang X, Zhao Q, Li Z, Fu F, Zhang H, Zheng M, Zhang S. Molecular Mechanism of Stem Cell Differentiation into Adipocytes and Adipocyte Differentiation of Malignant Tumor. Stem Cells Int 2020; 2020:8892300. [PMID: 32849880 PMCID: PMC7441422 DOI: 10.1155/2020/8892300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/07/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Adipogenesis is the process through which preadipocytes differentiate into adipocytes. During this process, the preadipocytes cease to proliferate, begin to accumulate lipid droplets, and develop morphologic and biochemical characteristics of mature adipocytes. Mesenchymal stem cells (MSCs) are a type of adult stem cells known for their high plasticity and capacity to generate mesodermal and nonmesodermal tissues. Many mature cell types can be generated from MSCs, including adipocyte, osteocyte, and chondrocyte. The differentiation of stem cells into multiple mature phenotypes is at the basis for tissue regeneration and repair. Cancer stem cells (CSCs) play a very important role in tumor development and have the potential to differentiate into multiple cell lineages. Accumulating evidence has shown that cancer cells can be induced to differentiate into various benign cells, such as adipocytes, fibrocytes, osteoblast, by a variety of small molecular compounds, which may provide new strategies for cancer treatment. Recent studies have reported that tumor cells undergoing epithelial-to-mesenchymal transition can be induced to differentiate into adipocytes. In this review, molecular mechanisms, signal pathways, and the roles of various biological processes in adipose differentiation are summarized. Understanding the molecular mechanism of adipogenesis and adipose differentiation of cancer cells may contribute to cancer treatments that involve inducing differentiation into benign cells.
Collapse
Affiliation(s)
- Kexin Zhang
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- 2Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Xudong Yang
- 3Tianjin Rehabilitation Center, Tianjin, China
| | - Qi Zhao
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Zugui Li
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- 4Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fangmei Fu
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- 4Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hao Zhang
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- 4Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Minying Zheng
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- 1Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
9
|
Liu X, Li X, Tao Y, Li N, Ji M, Zhang X, Chen Y, He Z, Yu K, Yu Z. TCDD inhibited the osteogenic differentiation of human fetal palatal mesenchymal cells through AhR and BMP-2/TGF-β/Smad signaling. Toxicology 2019; 431:152353. [PMID: 31887333 DOI: 10.1016/j.tox.2019.152353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022]
Abstract
Exposure to environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes cleft palate at high rates, but little is known about the underlying biological mechanisms. In the present study, we cultured osteoblasts from human fetal palate mesenchymal cells (hFPMCs) to explore the effects of TCDD on osteogenic differentiation. The results showed that TCDD significantly decreased cell proliferation, alkaline phosphatase (ALP) activity and calcium deposition. RNA analyses and protein detection demonstrated that TCDD downregulated a wide array of pro-osteogenic biomarkers. Further investigation of the underlying molecular mechanisms revealed that exposure to TCDD activated aryl hydrocarbon receptor (AhR) signaling and inhibited BMP-2/TGF-β1/Smad pathway molecules. The inactivation of AhR signaling using CRISPR/Cas9-mediated AhR deletion or by genetic siRNA knockdown significantly blocked the effects induced by TCDD, suggesting a critical role of AhR activation in the TCDD-mediated inhibition of hFPMC osteogenic differentiation. The cotreatment with TGF-β1 or BMP-2 and TCDD significantly relieved the activation of AhR and rescued the impairment of osteogenesis caused by TCDD. Taken together, our findings indicated that TCDD inhibited the osteogenic differentiation of hFPMCs via crosstalk between AhR and BMP-2/TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xiaozhuan Liu
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Li
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuchang Tao
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ning Li
- College of Food Science and Technology, Henan Agricultural University, China
| | - Mengmeng Ji
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiuli Zhang
- Division of Blood Vessel Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yao Chen
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhidong He
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Kailun Yu
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zengli Yu
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; School of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Lorthongpanich C, Thumanu K, Tangkiettrakul K, Jiamvoraphong N, Laowtammathron C, Damkham N, U-Pratya Y, Issaragrisil S. YAP as a key regulator of adipo-osteogenic differentiation in human MSCs. Stem Cell Res Ther 2019; 10:402. [PMID: 31852542 PMCID: PMC6921580 DOI: 10.1186/s13287-019-1494-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/13/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into several cell types, including cartilage, fat, and bone. As a common progenitor, MSC differentiation has to be tightly regulated to maintain the balance of their differentiation commitment. It has been reported that the decision process of MSCs into fat and bone cells is competing and reciprocal. Several factors have been suggested as critical factors that affect adipo-osteogenic decision, including melatonin and smad4. Yes-associated protein (YAP) is an important effector protein in the Hippo signaling pathway that acts as a transcriptional regulator by activating the transcription of the genes involved in cell proliferation and anti-apoptosis. The non-canonical role of YAP in regulating bone homeostasis by promoting osteogenesis and suppressing adipogenesis was recently demonstrated in a mouse model. However, it is unclear whether YAP is also crucial for modulating human MSC differentiation to fat and bone. METHODS The expression level of YAP during MSC differentiation was modulated using pharmaceutical molecule and genetic experiments through gain- and loss-of-function approaches. RESULTS We demonstrated for the first time that YAP has a non-canonical role in regulating the balance of adipo-osteogenic differentiation of human MSCs. The result from synchrotron radiation-based Fourier transform infrared (FTIR) microspectroscopy showed unique metabolic fingerprints generated from YAP-targeted differentiated cells that were clearly distinguished from non-manipulated control. CONCLUSIONS These results, thus, identify YAP as an important effector protein that regulates human MSC differentiation to fat and bone and suggests the use of FTIR microspectroscopy as a promising technique in stem cell research.
Collapse
Affiliation(s)
- Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Kanjana Thumanu
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, Thailand
| | - Kantpitchar Tangkiettrakul
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Nittaya Jiamvoraphong
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Nattaya Damkham
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yaowalak U-Pratya
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok, Thailand
| |
Collapse
|
11
|
Delgado-Enciso I, Valtierra-Alvarez J, Paz-Garcia J, Preciado-Ramirez J, Soriano-Hernandez AD, Mendoza-Hernandez MA, Guzman-Esquivel J, Cabrera-Licona A, Delgado-Enciso J, Cortes-Bazan JL, Rodriguez-Sanchez IP, Martinez-Fierro ML, Cabrera-Medina AO, Barajas-Saucedo CE, Paz-Michel B. Patient-reported health outcomes for severe knee osteoarthritis after conservative treatment with an intra-articular cell-free formulation for articular cartilage regeneration combined with usual medical care vs. usual medical care alone: A randomized controlled trial. Exp Ther Med 2019; 17:3351-3360. [PMID: 30988711 PMCID: PMC6447772 DOI: 10.3892/etm.2019.7384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a major public health problem characterized by joint pain, fatigue, functional limitation and decreased quality of life of the patient, which results in increased use of healthcare services and high economical costs. A promising novel bioactive cell-free formulation (BIOF2) for cartilage regeneration has recently been tested in pre-clinical and clinical trials, and has demonstrated a success rate similar to that of total joint arthroplasty for the treatment of severe knee OA. The present study evaluated the efficacy of treatment with BIOF2, by including it within a conservative regimen of 'usual medical care' of knee OA, and whether its efficacy was affected in subgroups of patients presenting with comorbidities that exacerbate OA. A prospective, randomized, 2-arm parallel group phase III clinical trial was conducted, which included 105 patients in the 'usual medical care' group (paracetamol/NSAIDs and general care provided by the family physician) and 107 patients in the BIOF2 group (usual medical care + intra-articular BIOF2 application at 0, 1 and 2 months). Two aspects were evaluated at 0, 6 and 12 months: i) Minimal clinically important improvement (MCII), based on 30% improvement of pain from the baseline; and ii) the Patient Acceptable Symptom State (PASS), a questionnaire that determines patient well-being thresholds for articular pain and function. Adverse effects and regular NSAID use were registered. At 12 months, BIOF-2 treatment produced MCII in 70% of the patients and >50% achieved PASS. Excluding the patients with class 2 obesity or malalignment conditions (genu varum or genu valgum >20 degrees), the experimental treatment produced MCII and PASS in 100 and 92% of patients, respectively, compared with 25 and 8% in the group of usual medical care (P<0.001). No patient with malalignment and treatment with BIOF2 achieved PASS. Notably, there were no serious adverse effects. To conclude, BIOF2 is a safe therapeutic alternative that is easy to implement together with usual medical care for knee OA. Trial registration: Cuban Public Registry of Clinical Trials (RPCEC) Database RPCEC00000277. Retrospectively registered June, 2018.
Collapse
Affiliation(s)
- Ivan Delgado-Enciso
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Jose Valtierra-Alvarez
- Department of Traumatology, University Regional Hospital, Colima State Health Services, Colima 28019, Mexico
| | - Juan Paz-Garcia
- Department of Traumatology, Union Hospital Center, Villa de Alvarez, Colima 28970, Mexico
| | - Jorge Preciado-Ramirez
- Department of Traumatology, University Regional Hospital, Colima State Health Services, Colima 28019, Mexico
| | - Alejandro D. Soriano-Hernandez
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | | | - Jose Guzman-Esquivel
- Department of Research, General Hospital of Zone No. 1 IMSS, Villa de Alvarez, Colima 28983, Mexico
| | - Ariana Cabrera-Licona
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
| | - Josuel Delgado-Enciso
- Department of Research, Foundation for Cancer Ethics, Education and Research of The Cancerology State Institute, Colima 28085, Mexico
| | - Jose L. Cortes-Bazan
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
| | - Iram P. Rodriguez-Sanchez
- Department of Cellular Biology, School of Biological Sciences, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon 64460, Mexico
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Autonomous University of Zacatecas, Zacatecas 98160, Mexico
| | - Ana O. Cabrera-Medina
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Carlos E. Barajas-Saucedo
- Department of Research, Cancerology State Institute, Colima State Health Services, Colima 28000, Mexico
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Brenda Paz-Michel
- Department of Research, Esteripharma Mexico, Mexico City 03100, Mexico
| |
Collapse
|
12
|
Rapid evolution of a retro-transposable hotspot of ovine genome underlies the alteration of BMP2 expression and development of fat tails. BMC Genomics 2019; 20:261. [PMID: 30940097 PMCID: PMC6445056 DOI: 10.1186/s12864-019-5620-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Background Sheep have developed the ability to store fat in their tails, which is a unique way of reserving energy to survive a harsh environment. However, the mechanism underlying this adaptive trait remains largely unsolved. Results In the present study, we provide evidence for the genetic determinants of fat tails, based on whole genome sequences of 89 individual sheep. A genome-wide scan of selective sweep identified several candidate loci including a region at chromosome 13, a haplotype of which underwent rapid evolution and spread through fat-tailed populations in China and the Middle East. Sequence analysis revealed an inter-genic origin of this locus, which later became a hotspot of ruminant-specific retro-transposon named BovB. Additionally, the candidate locus was validated based on a fat- and thin-tailed cross population. The expression of an upstream gene BMP2 was differentially regulated between fat-tailed and thin-tailed individuals in tail adipose and several other tissue types. Conclusions Our findings suggest the fixation of fat tails in domestic sheep is caused by a selective sweep near a retro-transposable hotspot at chromosome 13, the diversity of which specifically affects the expression of BMP2. The present study has shed light onto the understanding of fat metabolism. Electronic supplementary material The online version of this article (10.1186/s12864-019-5620-6) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Park JS, Kim M, Song NJ, Kim JH, Seo D, Lee JH, Jung SM, Lee JY, Lee J, Lee YS, Park KW, Park SH. A Reciprocal Role of the Smad4-Taz Axis in Osteogenesis and Adipogenesis of Mesenchymal Stem Cells. Stem Cells 2018; 37:368-381. [PMID: 30444564 PMCID: PMC7379966 DOI: 10.1002/stem.2949] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/26/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into mature cells of various cell types. Although the differentiation process of MSCs requires lineage-specific transcription factors, the exact molecular mechanism that determines MSCs differentiation is not clearly addressed. Here, we demonstrate a Smad4-Taz axis as a new intrinsic regulator for adipo-osteogenic differentiation of MSCs and show that this function of Smad4 is independent of the transforming growth factor-β signal. Smad4 directly bound to the Taz protein and facilitated nuclear localization of Taz through its nuclear localization signal. Nuclear retention of Taz by direct binding to Smad4 increased expression of osteogenic genes through enhancing Taz-runt-related transcription factor 2 (Runx2) interactions in the C3H10T1/2 MSC cell line and preosteoblastic MC3T3-E1 cells, whereas it suppressed expression of adipogenic genes through promoting Taz-peroxisome proliferator-activated receptor-γ (PPARγ) interaction in C3H10T1/2 and preadipogenic 3T3-L1 cells. A reciprocal role of the Smad4 in osteogenic and adipogenic differentiation was also observed in human adipose tissue-derived stem cells (hASCs). Consequently, Smad4 depletion in C3H10T1/2 and hASCs reduced nuclear retention of Taz and thus caused the decreased interaction with Runx2 or PPARγ, resulting in delayed osteogenesis or enhanced adipogenesis of the MSC. Therefore, these findings provide insight into a novel function of Smad4 to regulate the balance of MSC lineage commitment through reciprocal targeting of the Taz protein in osteogenic and adipogenic differentiation pathways. Stem Cells 2019;37:368-381.
Collapse
Affiliation(s)
- Jin Seok Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Minbeom Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - No-Joon Song
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Jun-Hyeong Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Dongyeob Seo
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Ji-Hyung Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jae Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Jaewon Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Youn Sook Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Kye Won Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
14
|
Ji H, Ahn K, Cho H, Kim HE, Kim Y, Kim O. Sanguisorba officinalis L. extracts activate Wnt/β-catenin pathway, and subsequently control adipo-osteogenic differentiation. Biochem Biophys Res Commun 2018; 504:352-358. [DOI: 10.1016/j.bbrc.2018.08.196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 08/30/2018] [Indexed: 01/13/2023]
|
15
|
Kang ES, Kim DS, Suhito IR, Lee W, Song I, Kim TH. Two-dimensional material-based bionano platforms to control mesenchymal stem cell differentiation. Biomater Res 2018; 22:10. [PMID: 29619243 PMCID: PMC5879765 DOI: 10.1186/s40824-018-0120-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/09/2018] [Indexed: 12/20/2022] Open
Abstract
Background In the past decade, stem cells, with their ability to differentiate into various types of cells, have been proven to be resourceful in regenerative medicine and tissue engineering. Despite the ability to repair damaged parts of organs and tissues, the use of stem cells still entails several limitations, such as low differentiation efficiency and difficulties in guiding differentiation. To address these limitations, nanotechnology approaches have been recently implemented in stem cell research. It has been discovered that stem cells, in combination with carbon-based functional materials, show enhanced regenerative performances in varying biophysical conditions. In particular, several studies have reported solutions to the conventional quandaries in biomedical engineering, using synergetic effects of nanohybrid materials, as well as further development of technologies to recover from diverse health conditions such as bone fracture and strokes. Main text In this review, we discuss several prior studies regarding the application of various nanomaterials in controlling the behavior of stem cells. We focus on the potential of different types of nanomaterials, such as two-dimensional materials, gold nanoparticles, and three-dimensional nanohybrid composites, to control the differentiation of human mesenchymal stem cells (hMSCs). These materials have been found to affect stem cell functions via the adsorption of growth/differentiation factors on the surfaces of nanomaterials and the activation of signaling pathways that are mostly related to cell adhesion and differentiation (e.g., FAK, Smad, Erk, and Wnt). Conclusion Controlling stem cell differentiation using biophysical factors, especially the use of nanohybrid materials to functionalize underlying substrates wherein the cells attach and grow, is a promising strategy to achieve cells of interest in a highly efficient manner. We hope that this review will facilitate the use of other types of newly discovered and/or synthesized nanomaterials (e.g., metal transition dichalcogenides, non-toxic quantum dots, and metal oxide frameworks) for stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Ee-Seul Kang
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Da-Seul Kim
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Intan Rosalina Suhito
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Wanhee Lee
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Inbeom Song
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Tae-Hyung Kim
- 1School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea.,2Integrative Research Center for Two-Dimensional Functional Materials, Institute of Interdisciplinary Convergence Research, Chung-Ang University, Seoul, 06974 Republic of Korea
| |
Collapse
|
16
|
Combating Osteoarthritis through Stem Cell Therapies by Rejuvenating Cartilage: A Review. Stem Cells Int 2018; 2018:5421019. [PMID: 29765416 PMCID: PMC5885495 DOI: 10.1155/2018/5421019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Knee osteoarthritis (OA) is a chronic degenerative disorder which could be distinguished by erosion of articular cartilage, pain, stiffness, and crepitus. Not only aging-associated alterations but also the metabolic factors such as hyperglycemia, dyslipidemia, and obesity affect articular tissues and may initiate or exacerbate the OA. The poor self-healing ability of articular cartilage due to limited regeneration in chondrocytes further adversely affects the osteoarthritic microenvironment. Traditional and current surgical treatment procedures for OA are limited and incapable to reverse the damage of articular cartilage. To overcome these limitations, cell-based therapies are currently being employed to repair and regenerate the structure and function of articular tissues. These therapies not only depend upon source and type of stem cells but also on environmental conditions, growth factors, and chemical and mechanical stimuli. Recently, the pluripotent and various multipotent mesenchymal stem cells have been employed for OA therapy, due to their differentiation potential towards chondrogenic lineage. Additionally, the stem cells have also been supplemented with growth factors to achieve higher healing response in osteoarthritic cartilage. In this review, we summarized the current status of stem cell therapies in OA pathophysiology and also highlighted the potential areas of further research needed in regenerative medicine.
Collapse
|
17
|
Jevons LA, Houghton FD, Tare RS. Augmentation of musculoskeletal regeneration: role for pluripotent stem cells. Regen Med 2018; 13:189-206. [PMID: 29557248 DOI: 10.2217/rme-2017-0113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rise in the incidence of musculoskeletal diseases is attributed to an increasing ageing population. The debilitating effects of musculoskeletal diseases, coupled with a lack of effective therapies, contribute to huge financial strains on healthcare systems. The focus of regenerative medicine has shifted to pluripotent stem cells (PSCs), namely, human embryonic stem cells and human-induced PSCs, due to the limited success of adult stem cell-based interventions. PSCs constitute a valuable cell source for musculoskeletal regeneration due to their capacity for unlimited self-renewal, ability to differentiate into all cell lineages of the three germ layers and perceived immunoprivileged characteristics. This review summarizes methods for chondrogenic, osteogenic, myogenic and adipogenic differentiation of PSCs and their potential for therapeutic applications.
Collapse
Affiliation(s)
- Lauren A Jevons
- Centre for Human Development, Stem Cells & Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Franchesca D Houghton
- Centre for Human Development, Stem Cells & Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Rahul S Tare
- Centre for Human Development, Stem Cells & Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.,Department of Mechanical Engineering, Faculty of Engineering & the Environment, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
18
|
Enhanced chondrogenesis differentiation of human induced pluripotent stem cells by MicroRNA-140 and transforming growth factor beta 3 (TGFβ3). Biologicals 2018; 52:30-36. [DOI: 10.1016/j.biologicals.2018.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/07/2017] [Accepted: 01/26/2018] [Indexed: 12/17/2022] Open
|
19
|
Thiagarajan L, Abu‐Awwad HAM, Dixon JE. Osteogenic Programming of Human Mesenchymal Stem Cells with Highly Efficient Intracellular Delivery of RUNX2. Stem Cells Transl Med 2017; 6:2146-2159. [PMID: 29090533 PMCID: PMC5702512 DOI: 10.1002/sctm.17-0137] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/05/2017] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are being exploited in regenerative medicine due to their tri-lineage differentiation and immunomodulation activity. Currently, there are two major challenges when directing the differentiation of MSCs for therapeutic applications. First, chemical and growth factor strategies to direct osteogenesis in vivo lack specificity for targeted delivery with desired effects. Second, MSC differentiation by gene therapy is difficult as transfection with existing approaches is clinically impractical (viral transfection) or have low efficacy (lipid-mediated transfection). These challenges can be avoided by directly delivering nonvirally derived recombinant protein transcription factors with the glycosaminoglycan-binding enhanced transduction (GET) delivery system (P21 and 8R peptides). We used the osteogenic master regulator, RUNX2 as a programming factor due to its stage-specific role in osteochondral differentiation pathways. Herein, we engineered GET-fusion proteins and compared sequential osteogenic changes in MSCs, induced by exposure to GET fusion proteins or conventional stimulation methods (dexamethasone and Bone morphogenetic protein 2). By assessing loss of stem cell-surface markers, upregulation of osteogenic genes and matrix mineralization, we demonstrate that GET-RUNX2 efficiently transduces MSCs and triggers osteogenesis by enhancing target gene expression directly. The high transduction efficiency of GET system holds great promise for stem cell therapies by allowing reproducible transcriptional control in stem cells, potentially bypassing problems observed with high-concentration growth-factor or pleiotropic steroid therapies. Stem Cells Translational Medicine 2017;6:2146-2159.
Collapse
Affiliation(s)
- Lalitha Thiagarajan
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre of Biomolecular Sciences, School of PharmacyUniversity of NottinghamNottinghamUnited Kingdom
| | - Hosam Al‐Deen M. Abu‐Awwad
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre of Biomolecular Sciences, School of PharmacyUniversity of NottinghamNottinghamUnited Kingdom
| | - James E. Dixon
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre of Biomolecular Sciences, School of PharmacyUniversity of NottinghamNottinghamUnited Kingdom
| |
Collapse
|
20
|
McClelland Descalzo DL, Satoorian TS, Walker LM, Sparks NRL, Pulyanina PY, Zur Nieden NI. Glucose-Induced Oxidative Stress Reduces Proliferation in Embryonic Stem Cells via FOXO3A/β-Catenin-Dependent Transcription of p21(cip1). Stem Cell Reports 2017; 7:55-68. [PMID: 27411103 PMCID: PMC4945584 DOI: 10.1016/j.stemcr.2016.06.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 11/17/2022] Open
Abstract
Embryonic stem cells (ESCs), which are derived from a peri-implantation embryo, are routinely cultured in medium containing diabetic glucose (Glc) concentrations. While pregnancy in women with pre-existing diabetes may result in small embryos, whether such high Glc levels affect ESC growth remains uncovered. We show here that long-term exposure of ESCs to diabetic Glc inhibits their proliferation, thereby mimicking in vivo findings. Molecularly, Glc exposure increased oxidative stress and activated Forkhead box O3a (FOXO3a), promoting increased expression and activity of the ROS-removal enzymes superoxide dismutase and catalase and the cell-cycle inhibitors p21cip1 and p27kip1. Diabetic Glc also promoted β-catenin nuclear localization and the formation of a complex with FOXO3a that localized to the promoters of Sod2, p21cip1, and potentially p27kip1. Our results demonstrate an adaptive response to increases in oxidative stress induced by diabetic Glc conditions that promote ROS removal, but also result in a decrease in proliferation. Exposure of ESCs to diabetic glucose (Glc) induces oxidative stress ESCs fight oxidative stress via FOXO3a-mediated transcription of Sod2 FOXO3a activation promotes p21cip1 and p27kip1 expression and cell-cycle inhibition Glc regulates FOXO3a/β-catenin co-occupation of the p21 and Sod2 promoters
Collapse
Affiliation(s)
- Darcie L McClelland Descalzo
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - Tiffany S Satoorian
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - Lauren M Walker
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - Nicole R L Sparks
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - Polina Y Pulyanina
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - Nicole I Zur Nieden
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, 1113 Biological Sciences Building, Riverside, CA 92521, USA.
| |
Collapse
|
21
|
Moya A, Larochette N, Bourguignon M, El-Hafci H, Potier E, Petite H, Logeart-Avramoglou D. Osteogenic potential of adipogenic predifferentiated human bone marrow-derived multipotent stromal cells for bone tissue-engineering. J Tissue Eng Regen Med 2017; 12:e1511-e1524. [PMID: 28875591 DOI: 10.1002/term.2571] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 07/13/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
Abstract
In the present study, we evaluated the benefits of an adipogenic predifferentiation, the pathway most closely related to osteoblastogenesis, on the pro-osteogenic potential of human adult multipotent bone marrow stromal cells (hBMSCs), both in vitro and in vivo. Adipogenic differentiation of hBMSCs for 14 days resulted in a heterogeneous cell population from which the most adipogenic-committed cells were eliminated by their lack of readhesion ability. Our results provided evidence that the select adherent adipogenic differentiated hBMSCs (sAD+ cells) express a gene profile characteristic of both adipogenic and osteogenic lineages. In vitro, when cultured in osteogenic medium, sAD+ differentiated along the osteogenic lineage faster than undifferentiated hBMSCs. In vivo, in an ectopic mouse model, sAD+ exhibited a significantly higher bone formation capability compared with undifferentiated hBMSCs. We sought, then, to investigate the underlying mechanisms responsible for such beneficial effects of adipogenic predifferentiation on bone formation and found that this outcome was not linked to a better cell survival post-implantation. The secretome of sAD+ was both proangiogenic and chemoattractant, but its potential did not supersede the one of undifferentiated hBMSCs. However, using co-culture systems, we observed that the sAD+ paracrine factors were pro-osteogenic on undifferentiated hBMSCs. In conclusion, adipogenic priming endows hBMSCs with high osteogenic potential as well as pro-osteogenic paracrine-mediated activity. This preconditioning appears as a promising strategy for bone tissue engineering technology in order to improve the hBMSC osteogenic potency in vivo.
Collapse
Affiliation(s)
- Adrien Moya
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | - Hanane El-Hafci
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Esther Potier
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Hervé Petite
- UMR 7052 CNRS University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | |
Collapse
|
22
|
Mahboudi H, Soleimani M, Enderami SE, Kehtari M, Hanaee-Ahvaz H, Ghanbarian H, Bandehpour M, Nojehdehi S, Mirzaei S, Kazemi B. The effect of nanofibre-based polyethersulfone (PES) scaffold on the chondrogenesis of human induced pluripotent stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1948-1956. [PMID: 29103309 DOI: 10.1080/21691401.2017.1396998] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human induced pluripotent stem cells (iPSCs) have been shown to have promising potential for regenerative medicine and tissue engineering applications. Chondrogenic differentiation of iPSCs is important for application in cartilage tissue engineering. In this study, we considered the effect of nanofibre-based polyethersulfone (PES) scaffold on the chondrogenesis of iPSCs. IPSC cells were cultured on the PES scaffold and scaffold free method. After 21 d, real-time PCR was performed to evaluate the cartilage-specific genes in the mRNA levels. For confirm our results, we have done immunocytochemistry and scanning electron microscopy (SEM) imaging. According to the results, higher significant expressions of common chondrogenic-related genes such as aggrecan, collagen type II and collagen type X were observed in PES seeded human iPSCs when compared to the mRNA levels measured in scaffold free method. Expression of collagen type I down regulated in both methods. Also, both methods were showed a similar pattern of expression of SOX9. Our results showed that nanofibre-based PES scaffold enhanced the chondrogenesis of iPSCs and the highest capacity for differentiation into chondrocyte-like cells. These cells and PES scaffold were demonstrated to have great efficiency for treatment of cartilage damages and lesions.
Collapse
Affiliation(s)
- Hossein Mahboudi
- a Department of Biotechnology , School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Masoud Soleimani
- b Hematology Department, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | - Seyed Ehsan Enderami
- c Cancer Gene Therapy Research Center, Faculty of Medicine , Zanjan University of Medical Sciences , Zanjan , Iran.,d Department of Stem Cell Biology , Stem Cell Technology Research Center , Tehran , Iran
| | - Mousa Kehtari
- d Department of Stem Cell Biology , Stem Cell Technology Research Center , Tehran , Iran
| | - Hana Hanaee-Ahvaz
- d Department of Stem Cell Biology , Stem Cell Technology Research Center , Tehran , Iran
| | - Hossein Ghanbarian
- a Department of Biotechnology , School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mojgan Bandehpour
- a Department of Biotechnology , School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Shahrzad Nojehdehi
- d Department of Stem Cell Biology , Stem Cell Technology Research Center , Tehran , Iran
| | - Samaneh Mirzaei
- d Department of Stem Cell Biology , Stem Cell Technology Research Center , Tehran , Iran
| | - Bahram Kazemi
- e Cellular and Molecular Biology Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
23
|
Sallustio F, Curci C, Aloisi A, Toma CC, Marulli E, Serino G, Cox SN, De Palma G, Stasi A, Divella C, Rinaldi R, Schena FP. Inhibin-A and Decorin Secreted by Human Adult Renal Stem/Progenitor Cells Through the TLR2 Engagement Induce Renal Tubular Cell Regeneration. Sci Rep 2017; 7:8225. [PMID: 28811645 PMCID: PMC5557965 DOI: 10.1038/s41598-017-08474-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
Acute kidney injury (AKI) is a public health problem worldwide. Several therapeutic strategies have been made to accelerate recovery and improve renal survival. Recent studies have shown that human adult renal progenitor cells (ARPCs) participate in kidney repair processes, and may be used as a possible treatment to promote regeneration in acute kidney injury. Here, we show that human tubular ARPCs (tARPCs) protect physically injured or chemically damaged renal proximal tubular epithelial cells (RPTECs) by preventing cisplatin-induced apoptosis and enhancing proliferation of survived cells. tARPCs without toll-like receptor 2 (TLR2) expression or TLR2 blocking completely abrogated this regenerative effect. Only tARPCs, and not glomerular ARPCs, were able to induce tubular cell regeneration process and it occurred only after damage detection. Moreover, we have found that ARPCs secreted inhibin-A and decorin following the RPTEC damage and that these secreted factors were directly involved in cell regeneration process. Polysaccharide synthetic vesicles containing these molecules were constructed and co-cultured with cisplatin damaged RPTECs. These synthetic vesicles were not only incorporated into the cells, but they were also able to induce a substantial increase in cell number and viability. The findings of this study increase the knowledge of renal repair processes and may be the first step in the development of new specific therapeutic strategies for renal repair.
Collapse
Affiliation(s)
- Fabio Sallustio
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Claudia Curci
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Alessandra Aloisi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,Institute of Microelectronics and Microsystems (C.N.R.- I.M.M.), via Monteroni, Campus Ecotekne, 73100, Lecce, Italy
| | - Chiara Cristina Toma
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Elisabetta Marulli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Grazia Serino
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari, 70013, Italy
| | - Sharon Natasha Cox
- C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Giuseppe De Palma
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy.,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy
| | - Alessandra Stasi
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Chiara Divella
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Rosaria Rinaldi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Nanoscience, Via Arnesano 16, 73100, Lecce, Italy.,University of Salento, Mathematics and Physics "E. De Giorgi" Department, University of Salento, 73100, Lecce, Italy
| | - Francesco Paolo Schena
- University of Bari, Department of Emergency and Organ Transplantation, Piazza G. Cesare 11, 70124, Bari, Italy. .,C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy. .,Schena Foundation, Strada Prov. le Valenzano-Casamassima Km 3, 70100, Valenzano (Ba), Italy.
| |
Collapse
|
24
|
Schorn L, Sproll C, Ommerborn M, Naujoks C, Kübler NR, Depprich R. Vertical bone regeneration using rhBMP-2 and VEGF. Head Face Med 2017; 13:11. [PMID: 28592312 PMCID: PMC5463342 DOI: 10.1186/s13005-017-0146-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/29/2017] [Indexed: 11/25/2022] Open
Abstract
Background Sufficient vertical and lateral bone supply and a competent osteogenic healing process are prerequisities for the successful osseointegration of dental implants in the alveolar bone. Several techniques including autologous bone grafts and guided bone regeneration are applied to improve quality and quantity of bone at the implantation site. Depending on the amount of lacking bone one- or two-stage procedures are required. Vertical bone augmentation has proven to be a challenge particularly in terms of bone volume stability. This study focuses on the three dimensional vertical bone generation in a one stage procedure in vivo. Therefore, a collagenous disc-shaped scaffold (ICBM = Insoluble Collagenous Bone Matrix) containing rhBMP-2 (Bone Morphogenetic Protein-2) and/or VEGF (Vascular Endothelial Growth Factor) was applied around the coronal part of a dental implant during insertion. RhBMP-2 and VEGF released directly at the implantation site were assumed to induce the generation of new vertical bone around the implant. Methods One hundred eight titanium implants were inserted into the mandible and the tibia of 12 mini pigs. Four experimental groups were formed: Control group, ICBM, ICBM + BMP-2, and ICBM + BMP-2 + VEGF. After 1, 4 and 12 weeks the animals were sacrificed and bone generation was investigated histologically and histomorphometrically. Results After 12 weeks the combination of ICBM + rhBMP2 + VEGF showed significantly more bone volume density (BVD%), a higher vertical bone gain (VBG) and more vertical bone gain around the implant (PVBG) in comparison to the control group. Conclusion By using collagenous disc-shaped matrices in combination with rhBMP-2 and VEGF vertical bone can be generated in a one stage procedure without donor site morbidity. The results of the presenting study suggest that the combination of rhBMP-2 and VEGF applied locally by using a collagenous carrier improves vertical bone generation in vivo. Further research is needed to establish whether this technique is applicable in clinical routines.
Collapse
Affiliation(s)
- Lara Schorn
- Department of Oral-, Maxillo- and Plastic Facial Surgery, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Christoph Sproll
- Department of Oral-, Maxillo- and Plastic Facial Surgery, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Michelle Ommerborn
- Department of Operative and Preventive Dentistry and Endodontics, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, Duesseldorf, 40225, Germany
| | - Christian Naujoks
- Department of Oral-, Maxillo- and Plastic Facial Surgery, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Norbert R Kübler
- Department of Oral-, Maxillo- and Plastic Facial Surgery, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Rita Depprich
- Department of Oral-, Maxillo- and Plastic Facial Surgery, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| |
Collapse
|
25
|
Açil Y, Möller B, Wiltfang J, Fändrich F, Ungefroren H. Programmable cells of monocytic origin as a source of osteochondroprogenitors: Effect of growth factors on osteogenic differentiation. J Craniomaxillofac Surg 2017; 45:1515-1520. [PMID: 28688862 DOI: 10.1016/j.jcms.2017.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/02/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
We have demonstrated previously that peripheral blood monocytes can be converted in vitro to a multipotent stem cell-like cell termed programmable cell of monocytic origin (PCMO) and subsequently into cells with chondrocyte-like phenotype. Here, we investigated whether PCMO could also be differentiated into osteoblast-like cells using growth factors with known osteoinductive potency. Following stimulation with BMP-2, BMP-7, IGF-1 or TGF-β1 for 7 and 14 days, PCMOs were analyzed for mRNA expression of collagen types I and V, alkaline phosphatase, osteocalcin, runt-related transcription factor-2 (Runx2) and Osterix (Osx) by quantitative RT-PCR (qPCR) and the levels of collagen I in culture supernatants by ELISA. The expression of osteoblastic markers was evident, albeit at a different extent in cultures of PCMOs after treatment with the above-mentioned growth factors. Culture supernatants from PCMOs stimulated for 6-10 days with BMP-2, BMP-7, IGF-1 or TGF-β1 contained high levels of collagen type I, together with earlier data indicating synthesis and proper secretion. The findings suggest that PCMOs can transform into cells that are phenotypically similar to osteoblasts and identify these cells as osteochondroprogenitors. The possibility of differentiating PCMOs from peripheral blood in sizable quantities could be a novel way to obtain autologous bone-like substitutes without donor-site morbidity.
Collapse
Affiliation(s)
- Yahya Açil
- Clinic of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| | - Björn Möller
- Clinic of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| | - Jörg Wiltfang
- Clinic of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| | - Fred Fändrich
- Institute for Applied Cell Therapy, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| | - Hendrik Ungefroren
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
| |
Collapse
|
26
|
Blair HC, Larrouture QC, Li Y, Lin H, Beer-Stoltz D, Liu L, Tuan RS, Robinson LJ, Schlesinger PH, Nelson DJ. Osteoblast Differentiation and Bone Matrix Formation In Vivo and In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:268-280. [PMID: 27846781 DOI: 10.1089/ten.teb.2016.0454] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review the characteristics of osteoblast differentiation and bone matrix synthesis. Bone in air breathing vertebrates is a specialized tissue that developmentally replaces simpler solid tissues, usually cartilage. Bone is a living organ bounded by a layer of osteoblasts that, because of transport and compartmentalization requirements, produce bone matrix exclusively as an organized tight epithelium. With matrix growth, osteoblasts are reorganized and incorporated into the matrix as living cells, osteocytes, which communicate with each other and surface epithelium by cell processes within canaliculi in the matrix. The osteoblasts secrete the organic matrix, which are dense collagen layers that alternate parallel and orthogonal to the axis of stress loading. Into this matrix is deposited extremely dense hydroxyapatite-based mineral driven by both active and passive transport and pH control. As the matrix matures, hydroxyapatite microcrystals are organized into a sophisticated composite in the collagen layer by nucleation in the protein lattice. Recent studies on differentiating osteoblast precursors revealed a sophisticated proton export network driving mineralization, a gene expression program organized with the compartmentalization of the osteoblast epithelium that produces the mature bone matrix composite, despite varying serum calcium and phosphate. Key issues not well defined include how new osteoblasts are incorporated in the epithelial layer, replacing those incorporated in the accumulating matrix. Development of bone in vitro is the subject of numerous projects using various matrices and mesenchymal stem cell-derived preparations in bioreactors. These preparations reflect the structure of bone to variable extents, and include cells at many different stages of differentiation. Major challenges are production of bone matrix approaching the in vivo density and support for trabecular bone formation. In vitro differentiation is limited by the organization and density of osteoblasts and by endogenous and exogenous inhibitors.
Collapse
Affiliation(s)
- Harry C Blair
- 1 Veteran's Affairs Medical Center , Pittsburgh, Pennsylvania.,2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | - Yanan Li
- 3 Department of Stomatology, Chinese PLA General Hospital , Beijing, China
| | - Hang Lin
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Donna Beer-Stoltz
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Li Liu
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Rocky S Tuan
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lisa J Robinson
- 5 Department of Pathology, West Virginia University School of Medicine , Morgantown, West Virginia.,6 Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Paul H Schlesinger
- 7 Department of Cell Biology, Washington University , Saint Louis, Missouri
| | - Deborah J Nelson
- 8 Department of Neurobiology, Pharmacology & Physiology, University of Chicago , Chicago, Illinois
| |
Collapse
|
27
|
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: 15] [Impact Index Per Article: 1.9] [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
|
28
|
Gu Y, Zhou J, Wang Q, Fan W, Yin G. Ginsenoside Rg1 promotes osteogenic differentiation of rBMSCs and healing of rat tibial fractures through regulation of GR-dependent BMP-2/SMAD signaling. Sci Rep 2016; 6:25282. [PMID: 27141994 PMCID: PMC4855182 DOI: 10.1038/srep25282] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 04/14/2016] [Indexed: 11/09/2022] Open
Abstract
Fracture healing is closely related to the number and activity of bone marrow mesenchymal stem cells (BMSCs) near the fracture site. The present study was to investigate the effect of Rg1 on osteogenic differentiation of cultured BMSCs and related mechanisms and on the fracture healing in a fracture model. In vitro experiments showed that Rg1 promoted the proliferation and osteogenic differentiation of BMSCs. Western blot analyses demonstrated that Rg1 promoted osteogenic differentiation of BMSCs through the glucocorticoid receptor (GR)-dependent BMP-2/Smad signaling pathway. In vivo, X-ray examination showed that callus growth in rats treated with Rg1 was substantially faster than that in control rats after fracture. The results of H&E and Safranin-O/Fast Green staining revealed that, compared with controls, rats in the Rg1 treatment group had a significantly higher proportion of trabecular bone but a much lower proportion of fibers and cartilage components inside the callus. Micro-CT suggested that bone mineral density (BMD), percent bone volume (BV/TV), trabecular number (Tb.N), and trabecular thickness (Tb.Th) were significantly increased in the treatment group, whereas trabecular separation (Tb.Sp) was significantly reduced. Thus, Rg1 promotes osteogenic differentiation by activating the GR/BMP-2 signaling pathway, enhances bone calcification, and ultimately accelerates the fracture healing in rats.
Collapse
Affiliation(s)
- Yanqing Gu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing 210000, China
| | - Jinchun Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing 210000, China
| | - Qin Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing 210000, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing 210000, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guang Zhou Road, Nanjing 210000, China
| |
Collapse
|
29
|
Lee SE, Moon JJM, Kim EY, Park SP. Stem Cell-Derived Bioactive Materials Accelerate Development of Porcine In Vitro-Fertilized Embryos. Cell Reprogram 2016; 17:181-90. [PMID: 26053518 DOI: 10.1089/cell.2014.0110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells show the capability to proliferate in an undifferentiated state with long-term self-renewal, which gives the cells advantages for use as bioactive material (BM) for embryo culture in vitro. The objective of this experiment was to investigate the effect of two BMs-human adipose tissue-derived mesenchymal stem cell BM (hAT-MSC-BM) and human embryonic stem cell-derived BM (hESC-BM)-on porcine embryo development compared to commonly used bovine serum albumin (BSA) or serum treatment groups. In vitro-fertilized (IVF) embryos were cultured in PZM-5 with 4 mg/mL BSA until day 4 and equally divided into four groups. Starting from day 4 (until day 6), each group was treated with the following protein additives: 4 mg/mL BSA (control), 10% fetal bovine serum (FBS), 10% hAT-MSC-BM, or 10% hESC-BM. Our results show FBS- and two other BM-treated groups showed significant increases in blastocyst formation rate, hatching rate, and total cell number compared with the control group (p<0.05). The hAT-MSC-BM and hESC-BM treatment groups presented better-quality embryo development, especially from the middle expanding stage to hatching. In particular, the hAT-MSC-BM-treated group showed the highest developmental potential of all groups and formed the most expanding-stage blastocysts. The relative expression of reprogramming-related transcription factor (POU5F1, SOX2, DPPA5, and CDH1), antioxidant (PRDX5), and apoptosis (BCL2L1 and BIRC5) genes also increased in two types of BMs compared to the control. In addition, we investigated the protein synthesis of the tight junction- and gap junction-related genes, connexin 43 and zonula occludens-1 (ZO-1); these increased more than in the control. These results demonstrate that stem cell-derived BMs accelerate porcine preimplantation embryo development and that the BMs would be helpful in the development of preimplantation embryos.
Collapse
Affiliation(s)
- Seung-Eun Lee
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea
| | - Jeremiah Ji-Man Moon
- 2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea
| | - Eun-Young Kim
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea.,3 Mirae Cell Bio , Seoul 143-854, Korea
| | - Se-Pill Park
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea.,3 Mirae Cell Bio , Seoul 143-854, Korea
| |
Collapse
|
30
|
Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ 2016; 23:1128-39. [PMID: 26868907 PMCID: PMC4946886 DOI: 10.1038/cdd.2015.168] [Citation(s) in RCA: 807] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 11/03/2015] [Accepted: 12/01/2015] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs), a non-hematopoietic stem cell population first discovered in bone marrow, are multipotent cells capable of differentiating into mature cells of several mesenchymal tissues, such as fat and bone. As common progenitor cells of adipocytes and osteoblasts, MSCs are delicately balanced for their differentiation commitment. Numerous in vitro investigations have demonstrated that fat-induction factors inhibit osteogenesis, and, conversely, bone-induction factors hinder adipogenesis. In fact, a variety of external cues contribute to the delicate balance of adipo-osteogenic differentiation of MSCs, including chemical, physical, and biological factors. These factors trigger different signaling pathways and activate various transcription factors that guide MSCs to commit to either lineage. The dysregulation of the adipo-osteogenic balance has been linked to several pathophysiologic processes, such as aging, obesity, osteopenia, osteopetrosis, and osteoporosis. Thus, the regulation of MSC differentiation has increasingly attracted great attention in recent years. Here, we review external factors and their signaling processes dictating the reciprocal regulation between adipocytes and osteoblasts during MSC differentiation and the ultimate control of the adipo-osteogenic balance.
Collapse
|
31
|
Moseti D, Regassa A, Kim WK. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int J Mol Sci 2016; 17:ijms17010124. [PMID: 26797605 PMCID: PMC4730365 DOI: 10.3390/ijms17010124] [Citation(s) in RCA: 481] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 11/24/2022] Open
Abstract
Adipogenesis is the process by which precursor stem cells differentiate into lipid laden adipocytes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program. In mammalian cells, the peroxisome proliferator-activated receptor γ (PPARγ), and the CCAAT/enhancer binding proteins (C/EBPs) such as C/EBPα, β and δ are considered the key early regulators of adipogenesis, while fatty acid binding protein 4 (FABP4), adiponectin, and fatty acid synthase (FAS) are responsible for the formation of mature adipocytes. Excess accumulation of lipids in the adipose tissue leads to obesity, which is associated with cardiovascular diseases, type II diabetes and other pathologies. Thus, investigating adipose tissue development and the underlying molecular mechanisms is vital to develop therapeutic agents capable of curbing the increasing incidence of obesity and related pathologies. In this review, we address the process of adipogenic differentiation, key transcription factors and proteins involved, adipogenic regulators and potential anti-adipogenic bioactive molecules.
Collapse
Affiliation(s)
- Dorothy Moseti
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Alemu Regassa
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Woo-Kyun Kim
- Department of Poultry Science, University of Georgia, 303 Poultry Science Building, Athens, GA 30602-2772, USA.
| |
Collapse
|
32
|
Geng Y, Zhao Y, Schuster LC, Feng B, Lynn DA, Austin KM, Stoklosa JD, Morrison JD. A Chemical Biology Study of Human Pluripotent Stem Cells Unveils HSPA8 as a Key Regulator of Pluripotency. Stem Cell Reports 2015; 5:1143-1154. [PMID: 26549849 PMCID: PMC4682066 DOI: 10.1016/j.stemcr.2015.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 01/06/2023] Open
Abstract
Chemical biology methods such as high-throughput screening (HTS) and affinity-based target identification can be used to probe biological systems on a biomacromolecule level, providing valuable insights into the molecular mechanisms of those systems. Here, by establishing a human embryonal carcinoma cell-based HTS platform, we screened 171,077 small molecules for regulators of pluripotency and identified a small molecule, Displurigen, that potently disrupts hESC pluripotency by targeting heat shock 70-kDa protein 8 (HSPA8), the constitutively expressed member of the 70-kDa heat shock protein family, as elucidated using affinity-based target identification techniques and confirmed by loss-of-function and gain-of-function assays. We demonstrated that HSPA8 maintains pluripotency by binding to the master pluripotency regulator OCT4 and facilitating its DNA-binding activity.
Collapse
Affiliation(s)
- Yijie Geng
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yongfeng Zhao
- Stem Cell Center, Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Lisa Corinna Schuster
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Bradley Feng
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Dana A Lynn
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Katherine M Austin
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jason Daniel Stoklosa
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Joseph D Morrison
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
33
|
Chan KH, Zhuo S, Ni M. Priming the Surface of Orthopedic Implants for Osteoblast Attachment in Bone Tissue Engineering. Int J Med Sci 2015; 12:701-7. [PMID: 26392807 PMCID: PMC4571547 DOI: 10.7150/ijms.12658] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/14/2015] [Indexed: 01/04/2023] Open
Abstract
The development of better orthopedic implants is incessant. While current implants can function reliably in the human body for a long period of time, there are still a significant number of cases for which the implants can fail prematurely due to poor osseointegration of the implant with native bone. Increasingly, it is recognized that it is extremely important to facilitate the attachment of osteoblasts on the implant so that a proper foundation of extracellular matrix (ECM) can be laid down for the growth of new bone tissue. In order to facilitate the osseointegration of the implant, both the physical nanotopography and chemical functionalization of the implant surface have to be optimized. In this short review, however, we explore how simple chemistry procedures can be used to functionalize the surfaces of three major classes of orthopedic implants, i.e. ceramics, metals, and polymers, so that the attachment of osteoblasts on implants can be facilitated in order to promote implant osseointegration.
Collapse
Affiliation(s)
- Kiat Hwa Chan
- 2. Institute of Bioengineering and Nanotechnology, Nanos, Singapore 138669, Singapore
| | - Shuangmu Zhuo
- 1. Institute of Laser and Optoelectronics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Ming Ni
- 3. Institute of Bioengineering and Nanotechnology, Nanos, Singapore 138669, Singapore
| |
Collapse
|
34
|
Sallustio F, Serino G, Schena FP. Potential Reparative Role of Resident Adult Renal Stem/Progenitor Cells in Acute Kidney Injury. Biores Open Access 2015; 4:326-33. [PMID: 26309808 PMCID: PMC4509615 DOI: 10.1089/biores.2015.0011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human kidney is particularly susceptible to ischemia and toxins with consequential tubular necrosis and activation of inflammatory processes. This process can lead to the acute renal injury, and even if the kidney has a great capacity for regeneration after tubular damage, in several circumstances, the normal renal repair program may not be sufficient to achieve a successful regeneration. Resident adult renal stem/progenitor cells could participate in this repair process and have the potentiality to enhance the renal regenerative mechanism. This could be achieved both directly, by means of their capacity to differentiate and integrate into the renal tissues, and by means of paracrine factors able to induce or improve the renal repair or regeneration. Recent genetic fate-tracing studies indicated that tubular damage is instead repaired by proliferative duplication of epithelial cells, acquiring a transient progenitor phenotype and by fate-restricted clonal cell progeny emerging from different nephron segments. In this review, we discuss about the properties and the reparative characteristics of high regenerative CD133(+)/CD24(+) cells, with a view to a future application of these cells for the treatment of acute renal injury.
Collapse
Affiliation(s)
- Fabio Sallustio
- Department of Emergency and Organ Transplantation, University of Bari , Bari, Italy . ; C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3 , Valenzano, Italy . ; Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento , Lecce-Monteroni, Lecce, Italy
| | - Grazia Serino
- Department of Emergency and Organ Transplantation, University of Bari , Bari, Italy
| | - Francesco Paolo Schena
- C.A.R.S.O. Consortium, Strada Prov. le Valenzano-Casamassima Km 3 , Valenzano, Italy . ; Schena Foundation, Research Center of Renal Diseases , Bari, Italy
| |
Collapse
|
35
|
zur Nieden NI, Turgman CC, Lang X, Larsen JM, Granelli J, Hwang YJ, Lyubovitsky JG. Fluorescent hydrogels for embryoid body formation and osteogenic differentiation of embryonic stem cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:10599-10605. [PMID: 25905907 DOI: 10.1021/acsami.5b02368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Substrate mechanics (e.g., stiffness and topography of the microenvironment) are likely critical for driving normal morphogenesis and tissue development. As such, substrate mechanics imposed by hydrogels have been exploited to guide the lineage differentiation of stem cells and to drive stemness. In this work, we chemically modified gelatin hydrogels through glyceraldehyde cross-linking to render them suitable for cell culture. The modified hydrogels proved to be ideal for embryonic stem cell osteogenesis, initially providing a soft nonadhesive surface for the formation of embryoid bodies. They subsequently degraded in culture to afford a harder surface during osteoblast differentiation. The gels synthesized are highly fluorescent, relatively easy to prepare, and can potentially aid in overcoming the challenge of imaging changes to the microenvironments of cells during three-dimensional cell culture. Exploiting these materials could lead to the development of tissue-engineered products of increased complexity and rational treatment strategies.
Collapse
|
36
|
Ehnes DD, Geransar RM, Rancourt DE, Zur Nieden NI. Exogenous nitric oxide enhances calcification in embryonic stem cell-derived osteogenic cultures. Differentiation 2015; 89:97-103. [PMID: 25929821 DOI: 10.1016/j.diff.2015.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 12/17/2022]
Abstract
While the involvement of nitric oxide in bone formation, homeostasis and healing has been extensively characterized, its role in directing pluripotent stem cells to the osteogenic lineage has not been described. Yet, the identification of chemical inducers that improve differentiation output to a particular lineage is highly valuable to the development of such cells for the cell-based treatment of osteo-degenerative diseases. This study aimed at investigating the instructive role of nitric oxide (NO) and its synthesizing enzymes on embryonic stem cell (ESC) osteogenic differentiation. Our findings showed that NO levels may support osteogenesis, but that the effect of nitric oxide on osteoblast differentiation may be specific to a particular time phase during the development of osteoblasts in vitro. Endogenously, nitric oxide was specifically secreted by osteogenic cultures during the calcification period. Simultaneously, messenger RNAs for both the endothelial and inducible nitric oxide synthase isoforms (eNOS and iNOS) were upregulated during this late phase development. However, the specific eNOS inhibitor L-N(5)-(1-Iminoethyl)ornithine dihydrochloride attenuated calcification more so than the specific iNOS inhibitor diphenyleneiodonium. Exogenous stage-specific supplementation of culture medium with the NO donor S-nitroso-N-acetyl-penicillamine increased the percentage of cells differentiating into osteoblasts and enhanced calcification. Our results point to a primary role for eNOS as a pro-osteogenic trigger in ESC differentiation and expand on the variety of supplements that may be used to direct ESC fate to the osteogenic lineage, which will be important in the development of cell-based therapies for osteo-degenerative diseases.
Collapse
Affiliation(s)
- D D Ehnes
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - R M Geransar
- Institute of Maternal & Child Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1
| | - D E Rancourt
- Institute of Maternal & Child Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1
| | - N I Zur Nieden
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA; Institute of Maternal & Child Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1.
| |
Collapse
|
37
|
Ehnes DD, Price FD, Shrive NG, Hart DA, Rancourt DE, zur Nieden NI. Embryonic stem cell-derived osteocytes are capable of responding to mechanical oscillatory hydrostatic pressure. J Biomech 2015; 48:1915-21. [PMID: 25936968 DOI: 10.1016/j.jbiomech.2015.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/04/2015] [Accepted: 04/08/2015] [Indexed: 11/28/2022]
Abstract
Osteoblasts can be derived from embryonic stem cells (ESCs) by a 30 day differentiation process, whereupon cells spontaneously differentiate upon removal of LIF and respond to exogenously added 1,25α(OH)2 vitamin D3 with enhanced matrix mineralization. However, bone is a load-bearing tissue that has to perform under dynamic pressure changes during daily movement, a capacity that is executed by osteocytes. At present, it is unclear whether ESC-derived osteogenic cultures contain osteocytes and whether these are capable of responding to a relevant cyclic hydrostatic compression stimulus. Here, we show that ESC-osteoblastogenesis is followed by the generation of osteocytes and then mechanically load ESC-derived osteogenic cultures in a compression chamber using a cyclic loading protocol. Following mechanical loading of the cells, iNOS mRNA was upregulated 31-fold, which was consistent with a role for iNOS as an immediate early mechanoresponsive gene. Further analysis of matrix and bone-specific genes suggested a cellular response in favor of matrix remodeling. Immediate iNOS upregulation also correlated with a concomitant increase in Ctnnb1 and Tcf7l2 mRNAs along with increased nuclear TCF transcriptional activity, while the mRNA for the repressive Tcf7l1 was downregulated, providing a possible mechanistic explanation for the noted matrix remodeling. We conclude that ESC-derived osteocytes are capable of responding to relevant mechanical cues, at least such that mimic oscillatory compression stress, which not only provides new basic understanding, but also information that likely will be important for their use in cell-based regenerative therapies.
Collapse
Affiliation(s)
- D D Ehnes
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - F D Price
- The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - N G Shrive
- McCaig Institute for Bone and Joint Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - D A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - D E Rancourt
- The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - N I zur Nieden
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA; The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
38
|
Yokoyama K, Ikeya M, Umeda K, Oda H, Nodomi S, Nasu A, Matsumoto Y, Izawa K, Horigome K, Kusaka T, Tanaka T, Saito MK, Yasumi T, Nishikomori R, Ohara O, Nakayama N, Nakahata T, Heike T, Toguchida J. Enhanced chondrogenesis of induced pluripotent stem cells from patients with neonatal-onset multisystem inflammatory disease occurs via the caspase 1-independent cAMP/protein kinase A/CREB pathway. Arthritis Rheumatol 2015; 67:302-14. [PMID: 25302486 DOI: 10.1002/art.38912] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 10/07/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Neonatal-onset multisystem inflammatory disease (NOMID) is a dominantly inherited autoinflammatory disease caused by NLRP3 mutations. NOMID pathophysiology is explained by the NLRP3 inflammasome, which produces interleukin-1β (IL-1β). However, epiphyseal overgrowth in NOMID is resistant to anti-IL-1 therapy and may therefore occur independently of the NLRP3 inflammasome. This study was undertaken to investigate the effect of mutated NLRP3 on chondrocytes using induced pluripotent stem cells (iPSCs) from patients with NOMID. METHODS We established isogenic iPSCs with wild-type or mutant NLRP3 from 2 NOMID patients with NLRP3 somatic mosaicism. The iPSCs were differentiated into chondrocytes in vitro and in vivo. The phenotypes of chondrocytes with wild-type and mutant NLRP3 were compared, particularly the size of the chondrocyte tissue produced. RESULTS Mutant iPSCs produced larger chondrocyte masses than wild-type iPSCs owing to glycosaminoglycan overproduction, which correlated with increased expression of the chondrocyte master regulator SOX9. In addition, in vivo transplantation of mutant cartilaginous pellets into immunodeficient mice caused disorganized endochondral ossification. Enhanced chondrogenesis was independent of caspase 1 and IL-1, and thus the NLRP3 inflammasome. Investigation of the human SOX9 promoter in chondroprogenitor cells revealed that the CREB/ATF-binding site was critical for SOX9 overexpression caused by mutated NLRP3. This was supported by increased levels of cAMP and phosphorylated CREB in mutant chondroprogenitor cells. CONCLUSION Our findings indicate that the intrinsic hyperplastic capacity of NOMID chondrocytes is dependent on the cAMP/PKA/CREB pathway, independent of the NLRP3 inflammasome.
Collapse
|
39
|
Mahmood A, Aldahmash A. Induction of primitive streak and mesendoderm formation in monolayer hESC culture by activation of TGF-β signaling pathway by Activin B. Saudi J Biol Sci 2015; 22:692-7. [PMID: 26586995 PMCID: PMC4625190 DOI: 10.1016/j.sjbs.2015.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/03/2015] [Indexed: 02/06/2023] Open
Abstract
Human embryonic stem cells (hESCs) have the ability to differentiate into all human cells, however controlling the differentiation has always been a challenge. In the present study we have investigated the direct differentiation of hESCs on MEFs by using TGF-β signaling pathway activators Activin A and Activin B. Activation of the TGF-β pathway with Activin B in low serum highly induced primitive streak and mesendoderm formation after 24 h, which included up-regulation of SOX 17 and BRACHYURY protein and gene expression. Continuous stimulation with Activin B in 2% serum further induced mesendoderm formation by increased gene expression of Brachyury, SOX17, MEOX and FOX at the same time we found down-regulation of neuroectodermal marker genes. Further, by stimulating the mesodermal cells by BMP-2 we succeeded to induce mesenchymal like cells with high expression of mesenchymal markers including; MEOX, FOX, RUNX2, COL1 and OSTEOPONTIN. In conclusion we have directed the differentiation of hESCs as monolayer to primitive streak like cells with Activin B and further into pure mesoderm and mesenchymal like cells by BMP-2.
Collapse
Affiliation(s)
- Amer Mahmood
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University and King Khalid University Hospital, Riyadh, Saudi Arabia
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University and King Khalid University Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Liu J, Nie H, Xu Z, Niu X, Guo S, Yin J, Guo F, Li G, Wang Y, Zhang C. The effect of 3D nanofibrous scaffolds on the chondrogenesis of induced pluripotent stem cells and their application in restoration of cartilage defects. PLoS One 2014; 9:e111566. [PMID: 25389965 PMCID: PMC4229072 DOI: 10.1371/journal.pone.0111566] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 10/05/2014] [Indexed: 11/19/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) rendered the reprogramming of terminally differentiated cells to primary stem cells with pluripotency possible and provided potential for the regeneration and restoration of cartilage defect. Chondrogenic differentiation of iPSCs is crucial for their application in cartilage tissue engineering. In this study we investigated the effect of 3D nanofibrous scaffolds on the chondrogenesis of iPSCs and articular cartilage defect restoration. Super-hydrophilic and durable mechanic polycaprolactone (PCL)/gelatin scaffolds were fabricated using two separate electrospinning processes. The morphological structure and mechanical properties of the scaffolds were characterized. The chondrogenesis of the iPSCs in vitro and the restoration of the cartilage defect was investigated using scanning electron microscopy (SEM), the Cell Counting Kit-8 (CCK-8), histological observation, RT-qPCR, and western blot analysis. iPSCs on the scaffolds expressed higher levels of chondrogenic markers than the control group. In an animal model, cartilage defects implanted with the scaffold-cell complex exhibited an enhanced gross appearance and histological improvements, higher cartilage-specific gene expression and protein levels, as well as subchondral bone regeneration. Therefore, we showed scaffolds with a 3D nanofibrous structure enhanced the chondrogenesis of iPSCs and that iPSC-containing scaffolds improved the restoration of cartilage defects to a greater degree than did scaffolds alone in vivo.
Collapse
Affiliation(s)
- Ji Liu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huarong Nie
- Institute of Polymers, School of Materials Science and Engineering, Nanchang University, Nanchang, China
| | - Zhengliang Xu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xin Niu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shangchun Guo
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junhui Yin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fei Guo
- Institute of Polymers, School of Materials Science and Engineering, Nanchang University, Nanchang, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regeneration Program, School of Biomedical Sciences, Room 904, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Yang Wang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (CZ); (YW)
| | - Changqing Zhang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (CZ); (YW)
| |
Collapse
|
41
|
Lorda-Diez CI, García-Porrero JA, Hurlé JM, Montero JA. Decorin gene expression in the differentiation of the skeletal connective tissues of the developing limb. Gene Expr Patterns 2014; 15:52-60. [DOI: 10.1016/j.gep.2014.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 11/28/2022]
|
42
|
Scimeca M, Giannini E, Antonacci C, Pistolese CA, Spagnoli LG, Bonanno E. Microcalcifications in breast cancer: an active phenomenon mediated by epithelial cells with mesenchymal characteristics. BMC Cancer 2014; 14:286. [PMID: 24758513 PMCID: PMC4021315 DOI: 10.1186/1471-2407-14-286] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 04/16/2014] [Indexed: 11/21/2022] Open
Abstract
Background Mammary microcalcifications have a crucial role in breast cancer detection, but the processes that induce their formation are unknown. Moreover, recent studies have described the occurrence of the epithelial–mesenchymal transition (EMT) in breast cancer, but its role is not defined. In this study, we hypothesized that epithelial cells acquire mesenchymal characteristics and become capable of producing breast microcalcifications. Methods Breast sample biopsies with microcalcifications underwent energy dispersive X-ray microanalysis to better define the elemental composition of the microcalcifications. Breast sample biopsies without microcalcifications were used as controls. The ultrastructural phenotype of breast cells near to calcium deposits was also investigated to verify EMT in relation to breast microcalcifications. The mesenchymal phenotype and tissue mineralization were studied by immunostaining for vimentin, BMP-2, β2-microglobulin, β-catenin and osteopontin (OPN). Results The complex formation of calcium hydroxyapatite was strictly associated with malignant lesions whereas calcium-oxalate is mainly reported in benign lesions. Notably, for the first time, we observed the presence of magnesium-substituted hydroxyapatite, which was frequently noted in breast cancer but never found in benign lesions. Morphological studies demonstrated that epithelial cells with mesenchymal characteristics were significantly increased in infiltrating carcinomas with microcalcifications and in cells with ultrastructural features typical of osteoblasts close to microcalcifications. These data were strengthened by the rate of cells expressing molecules typically involved during physiological mineralization (i.e. BMP-2, OPN) that discriminated infiltrating carcinomas with microcalcifications from those without microcalcifications. Conclusions We found significant differences in the elemental composition of calcifications between benign and malignant lesions. Observations of cell phenotype led us to hypothesize that under specific stimuli, mammary cells, which despite retaining a minimal epithelial phenotype (confirmed by cytokeratin expression), may acquire some mesenchymal characteristics transforming themselves into cells with an osteoblast-like phenotype, and are able to contribute to the production of breast microcalcifications.
Collapse
Affiliation(s)
| | | | | | | | | | - Elena Bonanno
- Anatomic Pathology Section, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy.
| |
Collapse
|
43
|
Fang X, Murakami H, Demura S, Hayashi K, Matsubara H, Kato S, Yoshioka K, Inoue K, Ota T, Shinmura K, Tsuchiya H. A novel method to apply osteogenic potential of adipose derived stem cells in orthopaedic surgery. PLoS One 2014; 9:e88874. [PMID: 24586422 PMCID: PMC3929506 DOI: 10.1371/journal.pone.0088874] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/16/2014] [Indexed: 01/22/2023] Open
Abstract
Background A number of publications have reported that adipose derived stem cells (ADSCs) have the capacity to be induced to differentiate into osteoblasts both in vitro and in vivo. However, it has been difficult to use separate ADSCs for cortical bone regeneration and bone reconstruction so far. Inspired by the research around stromal stem cells and cell sheets, we developed a new method to fabricate ADSCs sheets to accelerate and enhance the bone regeneration and bone reconstruction. Purpose To fabricate ADSCs sheets and evaluate their capacity to be induced to differentiate to osteoblasts in vitro. Methods Human adipose derived stem cells (hADSCs) were employed in this research. The fabricating medium containing 50 µM ascorbate-2-phosphate was used to enhance the secretion of collagen protein by the ADSCs and thus to make the cell sheets of ADSCs. As the separate ADSCs were divided into osteo-induction group and control group, the ADSCs sheets were also divided into two groups depending on induction by osteogenesis medium or no induction. The osteogenic capacity of each group was evaluated by ALP staining, Alizarin Red staining and ALP activity. Results The ADSCs sheets were fabricated after one-week culture in the fabricating medium. The ALP staining of ADSCs sheets showed positive results after 5 days osteo-induction and the Alizarin Red staining of ADSCs sheets showed positive results after 1 week osteo-induction. The ALP activity showed significant differences between these four groups. The ALP activity of ADSCs sheets groups showed higher value than that of separate ADSCs. Conclusion The experiments demonstrated that ADSCs sheets have better capacity than separate ADSCs to be induced to differentiate into osteoblasts. This indicates that it is possible to use the ADSCs sheets as a source of mesenchymal stem cells for bone regeneration and bone reconstruction.
Collapse
Affiliation(s)
- Xiang Fang
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Hideki Murakami
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Satoru Demura
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | | | - Satoshi Kato
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | | | - Kei Inoue
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Takashi Ota
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Kazuya Shinmura
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
- * E-mail:
| |
Collapse
|
44
|
Sidney LE, Kirkham GR, Buttery LD. Comparison of osteogenic differentiation of embryonic stem cells and primary osteoblasts revealed by responses to IL-1β, TNF-α, and IFN-γ. Stem Cells Dev 2014; 23:605-17. [PMID: 24192281 DOI: 10.1089/scd.2013.0336] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
There are well-established approaches for osteogenic differentiation of embryonic stem cells (ESCs), but few show direct comparison with primary osteoblasts or demonstrate differences in response to external factors. Here, we show comparative analysis of in vitro osteogenic differentiation of mouse ESC (osteo-mESC) and mouse primary osteoblasts. Both cell types formed mineralized bone nodules and produced osteogenic extracellular matrix, based on immunostaining for osteopontin and osteocalcin. However, there were marked differences in the morphology of osteo-mESCs and levels of mRNA expression for osteogenic genes. In response to the addition of proinflammatory cytokines interleukin-1β, tumor necrosis factor-α, and interferon-γ to the culture medium, primary osteoblasts showed increased production of nitric oxide (NO) and prostaglandin E2 (PGE2) at early time points and decreases in cell viability. In contrast, osteo-mESCs maintained viability and did not produce NO and PGE2 until day 21. The formation of bone nodules by primary osteoblasts was reduced markedly after cytokine stimulation but was unaffected in osteo-mESCs. Cell sorting of osteo-mESCs by cadherin-11 (cad-11) showed clear osteogenesis of cad-11(+) cells compared to unsorted osteo-mESCs and cad-11(-) cells. Moreover, the cad-11(+) cells showed a significant response to cytokines, similar to primary osteoblasts. Overall, these results show that while osteo-mESC cultures, without specific cell sorting, show characteristics of osteoblasts, there are also marked differences, notably in their responses to cytokine stimuli. These findings are relevant to understanding the differentiation of stem cells and especially developing in vitro models of disease, testing new drugs, and developing cell therapies.
Collapse
Affiliation(s)
- Laura E Sidney
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham , Nottingham, United Kingdom
| | | | | |
Collapse
|
45
|
Induced chondrogenic differentiation of parthenogenetic murine embryonic stem cells by insulin-like growth factor 2 treatment in a three-dimensional culture environment. Tissue Eng Regen Med 2013. [DOI: 10.1007/s13770-013-1100-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
46
|
Lee JK, Responte DJ, Cissell DD, Hu JC, Nolta JA, Athanasiou KA. Clinical translation of stem cells: insight for cartilage therapies. Crit Rev Biotechnol 2013; 34:89-100. [PMID: 24083452 DOI: 10.3109/07388551.2013.823596] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The limited regenerative capacity of articular cartilage and deficiencies of current treatments have motivated the investigation of new repair technologies. In vitro cartilage generation using primary cell sources is limited by cell availability and expansion potential. Pluripotent stem cells possess the capacity for chondrocytic differentiation and extended expansion, providing a potential future solution to cell-based cartilage regeneration. However, despite successes in producing cartilage using adult and embryonic stem cells, the translation of these technologies to the clinic has been severely limited. This review discusses recent advances in stem cell-based cartilage tissue engineering and the major current limitations to clinical translation of these products. Concerns regarding appropriate animal models and studies, stem cell manufacturing, and relevant regulatory processes and guidelines will be addressed. Understanding the significant hurdles limiting the clinical use of stem cell-based cartilage may guide future developments in the fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jennifer K Lee
- Department of Biomedical Engineering, University of California , Davis, CA , USA
| | | | | | | | | | | |
Collapse
|
47
|
Sritanaudomchai H, Kitiyanant Y, Tong-ngam P, Thonabulsombat C, White KL, Kusamran T. Enhanced chondrogenesis through specific growth factors in a buffalo embryonic stem cell model. Cell Biol Int 2013; 37:1246-58. [PMID: 23852953 DOI: 10.1002/cbin.10153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/08/2013] [Indexed: 11/10/2022]
Abstract
Chondrogenic differentiation of embryonic stem cells (ESCs) via embryoid bodies (EBs) is an established model to investigate chondrogenesis signaling pathways and molecular mechanisms in vitro. Our aim has been to improve upon the number of differentiated cells needed for the in vitro development of functional cartilage. Chondrogenic differentiation of buffalo ESCs was modulated by bone morphogenetic protein 2 (BMP-2), fibroblast growth factor 10 (FGF-10), transforming growth factor-beta1 (TGF-β1 ) individually and their combination. ESCs differentiation into chondrocytes was characterized by the appearance of Alcian blue-stained nodules and the expression of cartilage-associated genes (RT-PCR) and protein (immunocytochemistry). BMP-2 or FGF-10 treatment enhanced chondrogenic differentiation, whereas TGF-β1 treatment inhibited buffalo ESC-derived chondrogenesis. The combination of BMP-2 and FGF-10 was the most effective treatment. This treatment resulted in a higher number of Alcian blue-positive nodules by 15.2-fold, expression of the mesenchymal cell marker scleraxis gene by 3.25-fold, and the cartilage matrix protein collagen II gene and protein 1.9- and 7-fold, respectively, compared to the untreated control group. Chondrogenesis was also recapitulated from mesenchymal and chondrogenic progenitor cells, resulting in the establishment of mature chondrocytes. Thus, buffalo ESCs can be successfully triggered in vitro to differentiate into chondrocyte-like cells by specific growth factors, which may provide a novel in vitro model for further investigation of the regulatory mechanism(s) involved.
Collapse
Affiliation(s)
- Hathaitip Sritanaudomchai
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
48
|
Craft AM, Ahmed N, Rockel JS, Baht GS, Alman BA, Kandel RA, Grigoriadis AE, Keller GM. Specification of chondrocytes and cartilage tissues from embryonic stem cells. Development 2013; 140:2597-610. [PMID: 23715552 DOI: 10.1242/dev.087890] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.
Collapse
Affiliation(s)
- April M Craft
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, M5G 1L7, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kingham E, White K, Gadegaard N, Dalby MJ, Oreffo ROC. Nanotopographical cues augment mesenchymal differentiation of human embryonic stem cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:2140-2151. [PMID: 23362187 DOI: 10.1002/smll.201202340] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/01/2012] [Indexed: 06/01/2023]
Abstract
The production of bone-forming osteogenic cells for research purposes or transplantation therapies remains a significant challenge. Using planar polycarbonate substrates lacking in topographical cues and substrates displaying a nanotopographical pattern, mesenchymal differentiation of human embryonic stem cells is directed in the absence of chemical factors and without induction of differentiation by embryoid body formation. Cells incubated on nanotopographical substrates show enhanced expression of mesenchymal or stromal markers and expression of early osteogenic progenitors at levels above those detected in cells on planar substrates in the same basal media. Evidence of epithelial-to-mesenchymal transition during substrate differentiation and DNA methylation changes akin to chemical induction are also observed. These studies provide a suitable approach to overcome regenerative medical challenges and describe a defined, reproducible platform for human embryonic stem cell differentiation.
Collapse
Affiliation(s)
- Emmajayne Kingham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | | | | | | | | |
Collapse
|
50
|
Jian CX, Liu XF, Hu J, Li CJ, Zhang G, Li Y, Zhu JW, Tan YH. 20-Hydroxyecdysone-induced bone morphogenetic protein-2-dependent osteogenic differentiation through the ERK pathway in human periodontal ligament stem cells. Eur J Pharmacol 2013; 698:48-56. [PMID: 23397605 DOI: 10.1016/j.ejphar.2012.07.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
20-Hydroxyecdysone, an ecdysteroid hormone, can induce osteogenic differentiation in mesenchymal stem cells. Periodontal ligament stem cells (PDLS cells) have mesenchymal-stem-cell-like qualities and are considered as one of the candidates of future clinical application in periodontitis treatment. However, there are no studies describing the effect of 20-Hydroxyecdysone on PDLS cells. In this paper, we report a detailed study on the effect of 20-Hydroxyecdysone on PDLS cell proliferation in vitro. PDLS cells were developed from human PDL cells and were treated with 20-Hydroxyecdysone to understand different aspects of its effects. 20-Hydroxyecdysone promoted PDLS cell proliferation; significantly increased the gene expression levels of runt-related transcription factor 2, alkaline phosphatase (ALP), type I collagen, and osteocalcin. Moreover, 20-Hydroxyecdysone enhanced bone formation by PDLS cells and significantly increased bone morphogenetic protein-2 (BMP-2) mRNA and protein expression. However, 20-Hydroxyecdysonemediated increase in ALP activity was blocked with a BMP-2-specific neutralizing antibody or with the antagonist noggin; and20-Hydroxyecdysone mediated induction of BMP-2 expression and increase of ALP activity were abolished by the extracellular regulated protein kinase (ERK) MAPK pathway inhibitor PD98059. 20-Hydroxyecdysone also increased the phosphorylation of ERK. These findings provide evidence to state that 20-Hydroxyecdysone stimulates cell proliferation and induces osteogenic differentiation through the induction of BMP-2 expression in PDLS cells. It also shows that the ERK pathway is involved in 20-Hydroxyecdysone induced BMP-2 expression and osteogenic differentiation. These results are suggesting its potential as a drug for periodontal regenerative therapy.
Collapse
Affiliation(s)
- Cong-Xiang Jian
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, PR China
| | | | | | | | | | | | | | | |
Collapse
|