1
|
Lammi MJ, Qu C. Regulation of Oxygen Tension as a Strategy to Control Chondrocytic Phenotype for Cartilage Tissue Engineering and Regeneration. Bioengineering (Basel) 2024; 11:211. [PMID: 38534484 DOI: 10.3390/bioengineering11030211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Cartilage defects and osteoarthritis are health problems which are major burdens on health care systems globally, especially in aging populations. Cartilage is a vulnerable tissue, which generally faces a progressive degenerative process when injured. This makes it the 11th most common cause of global disability. Conservative methods are used to treat the initial phases of the illness, while orthopedic management is the method used for more progressed phases. These include, for instance, arthroscopic shaving, microfracturing and mosaicplasty, and joint replacement as the final treatment. Cell-based implantation methods have also been developed. Despite reports of successful treatments, they often suffer from the non-optimal nature of chondrocyte phenotype in the repair tissue. Thus, improved strategies to control the phenotype of the regenerating cells are needed. Avascular tissue cartilage relies on diffusion for nutrients acquisition and the removal of metabolic waste products. A low oxygen content is also present in cartilage, and the chondrocytes are, in fact, well adapted to it. Therefore, this raises an idea that the regulation of oxygen tension could be a strategy to control the chondrocyte phenotype expression, important in cartilage tissue for regenerative purposes. This narrative review discusses the aspects related to oxygen tension in the metabolism and regulation of articular and growth plate chondrocytes and progenitor cell phenotypes, and the role of some microenvironmental factors as regulators of chondrocytes.
Collapse
Affiliation(s)
- Mikko J Lammi
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden
| | - Chengjuan Qu
- Department of Odontology, Umeå University, SE-90187 Umeå, Sweden
| |
Collapse
|
2
|
Ceranski AK, Carreño-Gonzalez MJ, Ehlers AC, Colombo MV, Cidre-Aranaz F, Grünewald TGP. Hypoxia and HIFs in Ewing sarcoma: new perspectives on a multi-facetted relationship. Mol Cancer 2023; 22:49. [PMID: 36915100 PMCID: PMC10010019 DOI: 10.1186/s12943-023-01750-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Hypoxia develops during the growth of solid tumors and influences tumoral activity in multiple ways. Low oxygen tension is also present in the bone microenvironment where Ewing sarcoma (EwS) - a highly aggressive pediatric cancer - mainly arises. Hypoxia inducible factor 1 subunit alpha (HIF-1-a) is the principal molecular mediator of the hypoxic response in cancer whereas EWSR1::FLI1 constitutes the oncogenic driver of EwS. Interaction of the two proteins has been shown in EwS. Although a growing body of studies investigated hypoxia and HIFs in EwS, their precise role for EwS pathophysiology is not clarified to date. This review summarizes and structures recent findings demonstrating that hypoxia and HIFs play a role in EwS at multiple levels. We propose to view hypoxia and HIFs as independent protagonists in the story of EwS and give a perspective on their potential clinical relevance as prognostic markers and therapeutic targets in EwS treatment.
Collapse
Affiliation(s)
- A Katharina Ceranski
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Martha J Carreño-Gonzalez
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Anna C Ehlers
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Maria Vittoria Colombo
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Via F. Chiesa 5, CH-6500, Bellinzona, Switzerland.,Department of Surgery, Service of Orthopaedics and Traumatology, EOC, Lugano, Switzerland.,Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico Di Milano, Via Mancinelli 7, 20131, Milan, Italy
| | - Florencia Cidre-Aranaz
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Thomas G P Grünewald
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany. .,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
3
|
Ranmuthu CKI, Ranmuthu CDS, Wijewardena CK, Seah MKT, Khan WS. Evaluating the Effect of Hypoxia on Human Adult Mesenchymal Stromal Cell Chondrogenesis In Vitro : A Systematic Review. Int J Mol Sci 2022; 23:ijms232315210. [PMID: 36499531 PMCID: PMC9741425 DOI: 10.3390/ijms232315210] [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: 09/26/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Human adult mesenchymal stromal cells (MSCs) from a variety of sources may be used to repair defects in articular cartilage by inducing them into chondrogenic differentiation. The conditions in which optimal chondrogenic differentiation takes place are an area of interest in the field of tissue engineering. Chondrocytes exist in vivo in a normally hypoxic environment and thus it has been suggested that exposing MSCs to hypoxia may also contribute to a beneficial effect on their differentiation. There are two main stages in which MSCs can be exposed to hypoxia, the expansion phase when cells are cultured, and the differentiation phase when cells are induced with a chondrogenic medium. This systematic review sought to explore the effect of hypoxia at these two stages on human adult MSC chondrogenesis in vitro. A literature search was performed on PubMed, EMBASE, Medline via Ovid, and Cochrane, and 24 studies were ultimately included. The majority of these studies showed that hypoxia during the expansion phase or the differentiation phase enhances at least some markers of chondrogenic differentiation in adult MSCs. These results were not always demonstrated at the protein level and there were also conflicting reports. Studies evaluating continuous exposure to hypoxia during the expansion and differentiation phases also had mixed results. These inconsistent results can be explained by the heterogeneity of studies, including factors such as different sources of MSCs used, donor variability, level of hypoxia used in each study, time exposed to hypoxia, and differences in culture methodology.
Collapse
|
4
|
Rhatomy S, Setyawan R, Romulo MA. Enhancement of Chondrogenesis in Hypoxic Precondition Culture: A Systematic Review. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Cartilage tear has begun to be treated with stem cells. However, stem cell oxygen level culture has not been evaluated for the best environment to enhance chondrogenesis.
AIM: The purpose of this review is to focus on the hypoxic oxygen level of stem cells culture as a treatment for cartilage tear.
METHODS: A literature search was systemically conducted on PubMed (MEDLINE), OVID, EMBASE, the Cochrane Library, Scopus, Web of Science, Science Direct, Wiley Online Library, Google Scholar, and bibliography of selected articles with the terms (“culture”) AND (“stem cell” OR “mesenchymal stem cell” OR “MSC”) AND (“hypoxic” OR “hypoxia”) AND (“cartilage” OR “chondro*”) as the main keywords. A total of 438 articles were reviewed. Thirty-six articles were considered relevant for this systematic review.
RESULTS: The result of this review supports stimulation effects of hypoxic oxygen level stem cell culture in chondrogenesis process. Most studies used 5% oxygen concentration for culture, both of in vivo and in vitro studies. Due to the heterogeneity nature of the included studies, meta-analysis was unable to be conducted.
CONCLUSION: Hypoxia state seems to play an important role in chondrocytes proliferation, differentiation, and matrix production.
Collapse
|
5
|
Mohammadalipour A, Dumbali SP, Wenzel PL. Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy. Front Cell Dev Biol 2020; 8:603292. [PMID: 33365311 PMCID: PMC7750467 DOI: 10.3389/fcell.2020.603292] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cell (MSC) metabolism plays a crucial role in the surrounding microenvironment in both normal physiology and pathological conditions. While MSCs predominantly utilize glycolysis in their native hypoxic niche within the bone marrow, new evidence reveals the importance of upregulation in mitochondrial activity in MSC function and differentiation. Mitochondria and mitochondrial regulators such as sirtuins play key roles in MSC homeostasis and differentiation into mature lineages of the bone and hematopoietic niche, including osteoblasts and adipocytes. The metabolic state of MSCs represents a fine balance between the intrinsic needs of the cellular state and constraints imposed by extrinsic conditions. In the context of injury and inflammation, MSCs respond to reactive oxygen species (ROS) and damage-associated molecular patterns (DAMPs), such as damaged mitochondria and mitochondrial products, by donation of their mitochondria to injured cells. Through intercellular mitochondria trafficking, modulation of ROS, and modification of nutrient utilization, endogenous MSCs and MSC therapies are believed to exert protective effects by regulation of cellular metabolism in injured tissues. Similarly, these same mechanisms can be hijacked in malignancy whereby transfer of mitochondria and/or mitochondrial DNA (mtDNA) to cancer cells increases mitochondrial content and enhances oxidative phosphorylation (OXPHOS) to favor proliferation and invasion. The role of MSCs in tumor initiation, growth, and resistance to treatment is debated, but their ability to modify cancer cell metabolism and the metabolic environment suggests that MSCs are centrally poised to alter malignancy. In this review, we describe emerging evidence for adaptations in MSC bioenergetics that orchestrate developmental fate decisions and contribute to cancer progression. We discuss evidence and potential strategies for therapeutic targeting of MSC mitochondria in regenerative medicine and tissue repair. Lastly, we highlight recent progress in understanding the contribution of MSCs to metabolic reprogramming of malignancies and how these alterations can promote immunosuppression and chemoresistance. Better understanding the role of metabolic reprogramming by MSCs in tissue repair and cancer progression promises to broaden treatment options in regenerative medicine and clinical oncology.
Collapse
Affiliation(s)
- Amina Mohammadalipour
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sandeep P Dumbali
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pamela L Wenzel
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States.,Immunology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
6
|
Camacho-Cardenosa M, Camacho-Cardenosa A, Timón R, Olcina G, Tomas-Carus P, Brazo-Sayavera J. Can Hypoxic Conditioning Improve Bone Metabolism? A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16101799. [PMID: 31117194 PMCID: PMC6572511 DOI: 10.3390/ijerph16101799] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Among other functions, hypoxia-inducible factor plays a critical role in bone–vascular coupling and bone formation. Studies have suggested that hypoxic conditioning could be a potential nonpharmacological strategy for treating skeletal diseases. However, there is no clear consensus regarding the bone metabolism response to hypoxia. Therefore, this review aims to examine the impact of different modes of hypoxia conditioning on bone metabolism. The PubMed and Web of Science databases were searched for experimental studies written in English that investigated the effects of modification of ambient oxygen on bone remodelling parameters of healthy organisms. Thirty-nine studies analysed the effect of sustained or cyclic hypoxia exposure on genetic and protein expression and mineralisation capacity of different cell models; three studies carried out in animal models implemented sustained or cyclic hypoxia; ten studies examined the effect of sustained, intermittent or cyclic hypoxia on bone health and hormonal responses in humans. Different modes of hypoxic conditioning may have different impacts on bone metabolism both in vivo and in vitro. Additional research is necessary to establish the optimal cyclical dose of oxygen concentration and exposure time.
Collapse
Affiliation(s)
| | | | - Rafael Timón
- Faculty of Sport Science, University of Extremadura, 10003 Cáceres, Spain.
| | - Guillermo Olcina
- Faculty of Sport Science, University of Extremadura, 10003 Cáceres, Spain.
| | - Pablo Tomas-Carus
- Departamento de Desporto e Saúde, Escola de Ciência e Tecnologia, Universidade de Évora, 7000-812 Évora, Portugal.
- Comprehensive Health Research Centre (CHRC), University of Évora, 7000-812 Évora, Portugal.
| | - Javier Brazo-Sayavera
- Instituto Superior de Educación Física, Universidad de la República, 40000 Rivera, Uruguay.
- Polo de Desarrollo Universitario EFISAL, Universidad de la República, 40000 Rivera, Uruguay.
| |
Collapse
|
7
|
Zhou S, Chen S, Jiang Q, Pei M. Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis. Cell Mol Life Sci 2019; 76:1653-1680. [PMID: 30689010 PMCID: PMC6456412 DOI: 10.1007/s00018-019-03017-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/10/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Adult stem cells, also termed as somatic stem cells, are undifferentiated cells, detected among differentiated cells in a tissue or an organ. Adult stem cells can differentiate toward lineage specific cell types of the tissue or organ in which they reside. They also have the ability to differentiate into mature cells of mesenchymal tissues, such as cartilage, fat and bone. Despite the fact that the balance has been comprehensively scrutinized between adipogenesis and osteogenesis and between chondrogenesis and osteogenesis, few reviews discuss the relationship between chondrogenesis and adipogenesis. In this review, the developmental and transcriptional crosstalk of chondrogenic and adipogenic lineages are briefly explored, followed by elucidation of signaling pathways and external factors guiding lineage determination between chondrogenic and adipogenic differentiation. An in-depth understanding of overlap and discrepancy between these two mesenchymal tissues in lineage differentiation would benefit regeneration of high-quality cartilage tissues and adipose tissues for clinical applications.
Collapse
Affiliation(s)
- Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, 610083, Sichuan, People's Republic of China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- Robert C. Byrd Health Sciences Center, WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
8
|
Pattappa G, Johnstone B, Zellner J, Docheva D, Angele P. The Importance of Physioxia in Mesenchymal Stem Cell Chondrogenesis and the Mechanisms Controlling Its Response. Int J Mol Sci 2019; 20:E484. [PMID: 30678074 PMCID: PMC6387316 DOI: 10.3390/ijms20030484] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage covers the surface of synovial joints and enables joint movement. However, it is susceptible to progressive degeneration with age that can be accelerated by either previous joint injury or meniscectomy. This degenerative disease is known as osteoarthritis (OA) and it greatly affects the adult population. Cell-based tissue engineering provides a possible solution for treating OA at its earliest stages, particularly focal cartilage lesions. A candidate cell type for treating these focal defects are Mesenchymal Stem Cells (MSCs). However, present methods for differentiating these cells towards the chondrogenic lineage lead to hypertrophic chondrocytes and bone formation in vivo. Environmental stimuli that can stabilise the articular chondrocyte phenotype without compromising tissue formation have been extensively investigated. One factor that has generated intensive investigation in MSC chondrogenesis is low oxygen tension or physioxia (2⁻5% oxygen). In vivo articular cartilage resides at oxygen tensions between 1⁻4%, and in vitro results suggest that these conditions are beneficial for MSC expansion and chondrogenesis, particularly in suppressing the cartilage hypertrophy. This review will summarise the current literature regarding the effects of physioxia on MSC chondrogenesis with an emphasis on the pathways that control tissue formation and cartilage hypertrophy.
Collapse
Affiliation(s)
- Girish Pattappa
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| | - Johannes Zellner
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Denitsa Docheva
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Peter Angele
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
- Sporthopaedicum Regensburg, Hildegard von Bingen Strasse 1, 93053 Regensburg, Germany.
| |
Collapse
|
9
|
Wang W, Han ZC. Heterogeneity of Human Mesenchymal Stromal/Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1123:165-177. [DOI: 10.1007/978-3-030-11096-3_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
10
|
Andreeva ER, Matveeva DK. Multipotent Mesenchymal Stromal Cells and Extracellular Matrix: Regulation under Hypoxia. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s0362119718060038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Elkhenany H, AlOkda A, El-Badawy A, El-Badri N. Tissue regeneration: Impact of sleep on stem cell regenerative capacity. Life Sci 2018; 214:51-61. [PMID: 30393021 DOI: 10.1016/j.lfs.2018.10.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
The circadian rhythm orchestrates many cellular functions, such as cell division, cell migration, metabolism and numerous intracellular biological processes. The physiological changes during sleep are believed to promote a suitable microenvironment for stem cells to proliferate, migrate and differentiate. These effects are mediated either directly by circadian clock genes or indirectly via hormones and cytokines. Hormones, such as melatonin and cortisol, are secreted in response to neural optic signals and act in harmony to regulate many biological functions during sleep. Herein, we correlate the effects of the main circadian genes on the expression of certain stem cell genes responsible for the regeneration of different tissues, including bone, cartilage, skin, and intestine. We also review the effects of different hormones and cytokines on stem cell activation or suppression and their relationship to the day/night cycle. The correlation of circadian rhythm with tissue regeneration could have implications in understanding the biology of sleep and tissue regeneration and in enhancing the efficacy and timing of surgical procedures.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt; Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, 22785, Egypt
| | - Abdelrahman AlOkda
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Ahmed El-Badawy
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Nagwa El-Badri
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt.
| |
Collapse
|
12
|
Bae HC, Park HJ, Wang SY, Yang HR, Lee MC, Han HS. Hypoxic condition enhances chondrogenesis in synovium-derived mesenchymal stem cells. Biomater Res 2018; 22:28. [PMID: 30275971 PMCID: PMC6158840 DOI: 10.1186/s40824-018-0134-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Background The chondrogenic differentiation of mesenchymal stem cells (MSCs) is regulated by many factors, including oxygen tensions, growth factors, and cytokines. Evidences have suggested that low oxygen tension seems to be an important regulatory factor in the proliferation and chondrogenic differentiation in various MSCs. Recent studies report that synovium-derived mesenchymal stem cells (SDSCs) are a potential source of stem cells for the repair of articular cartilage defects. But, the effect of low oxygen tension on the proliferation and chondrogenic differentiation in SDSCs has not characterized. In this study, we investigated the effects of hypoxia on proliferation and chondrogenesis in SDSCs. Method SDSCs were isolated from patients with osteoarthritis at total knee replacement. To determine the effect of oxygen tension on proliferation and colony-forming characteristics of SDSCs, A colony-forming unit (CFU) assay and cell counting-based proliferation assay were performed under normoxic (21% oxygen) or hypoxic (5% oxygen). For in vitro chondrogenic differentiation, SDSCs were concentrated to form pellets and subjected to conditions appropriate for chondrogenic differentiation under normoxia and hypoxia, followed by the analysis for the expression of genes and proteins of chondrogenesis. qRT-PCR, histological assay, and glycosoaminoglycan assays were determined to assess chondrogenesis. Results Low oxygen condition significantly increased proliferation and colony-forming characteristics of SDSCs compared to that of SDSCs under normoxic culture. Similar pellet size and weight were found for chondrogensis period under hypoxia and normoxia condition. The mRNA expression of types II collagen, aggrecan, and the transcription factor SOX9 was increased under hypoxia condition. Histological sections stained with Safranin-O demonstrated that hypoxic conditions had increased proteoglycan synthesis. Immunohistochemistry for types II collagen demonstrated that hypoxic culture of SDSCs increased type II collagen expression. In addition, GAG deposition was significantly higher in hypoxia compared with normoxia at 21 days of differentiation. Conclusion These findings show that hypoxia condition has an important role in regulating the synthesis ECM matrix by SDSCs as they undergo chondrogenesis. This has important implications for cartilage tissue engineering applications of SDSCs.
Collapse
Affiliation(s)
- Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Hee Jung Park
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Sun Young Wang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Ha Ru Yang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Myung Chul Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| |
Collapse
|
13
|
MIF Plays a Key Role in Regulating Tissue-Specific Chondro-Osteogenic Differentiation Fate of Human Cartilage Endplate Stem Cells under Hypoxia. Stem Cell Reports 2017; 7:249-62. [PMID: 27509135 PMCID: PMC4982989 DOI: 10.1016/j.stemcr.2016.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/06/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022] Open
Abstract
Degenerative cartilage endplate (CEP) shows decreased chondrification and increased ossification. Cartilage endplate stem cells (CESCs), with the capacity for chondro-osteogenic differentiation, are responsible for CEP restoration. CEP is avascular and hypoxic, while the physiological hypoxia is disrupted in the degenerated CEP. Hypoxia promoted chondrogenesis but inhibited osteogenesis in CESCs. This tissue-specific differentiation fate of CESCs in response to hypoxia was physiologically significant with regard to CEP maintaining chondrification and refusing ossification. MIF, a downstream target of HIF1A, is involved in cartilage and bone metabolisms, although little is known about its regulatory role in differentiation. In CESCs, MIF was identified as a key point through which HIF1A regulated the chondro-osteogenic differentiation. Unexpectedly, unlike the traditionally recognized mode, increased nuclear-expressed MIF under hypoxia was identified to act as a transcriptional regulator by interacting with the promoter of SOX9 and RUNX2. This mode of HIF1A/MIF function may represent a target for CEP degeneration therapy. The hypoxic microenvironment is disrupted in degenerative CEP Hypoxia promotes chondrogenesis but inhibits osteogenesis in CESCs Hypoxia regulates chondro-osteogenesis through HIF1A/MIF pathway MIF acts as a transcriptional regulator under hypoxia
Collapse
|
14
|
Yao Y, Deng Q, Sun C, Song W, Liu H, Zhou Y. A genome-wide analysis of the gene expression profiles and alternative splicing events during the hypoxia-regulated osteogenic differentiation of human cartilage endplate-derived stem cells. Mol Med Rep 2017; 16:1991-2001. [PMID: 28656244 PMCID: PMC5562021 DOI: 10.3892/mmr.2017.6846] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 04/25/2017] [Indexed: 12/20/2022] Open
Abstract
It has been hypothesized that intervertebral disc degeneration is initiated by degeneration of the cartilage endplate (CEP), which is characterized by cartilage ossification. CEP‑derived stem cells (CESCs), with the potential for chondro‑osteogenic differentiation, may be responsible for the balance between chondrification and ossification in the CEP. The CEP remains in an avascular and hypoxic microenvironment; the present study observed that hypoxia was able to markedly inhibit the osteogenic differentiation of CESCs. This tissue‑specific CESC differentiation in response to a hypoxic microenvironment was physiologically important for the prevention of ossification in the CEP. In order to study the hypoxia‑regulated mechanisms underlying osteogenic differentiation of CESCs, a Human Transcriptome Array 2.0 was used to detect differentially expressed genes (DEGs) and alternatively spliced genes (ASGs) during the osteogenic differentiation of CESCs under hypoxia, compared with those induced under normoxia. High‑throughput analysis of DEGs and ASGs demonstrated that genes in the complement pathway were enriched, which may be a potential mechanism underlying hypoxia inhibition of CESCs osteogenesis. The results of the present study may provide a basis for future mechanistic studies regarding gene expression levels and alternative splicing events during the hypoxia‑regulated inhibition of osteogenesis, which may be helpful in identifying targets for CEP degeneration therapy.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Qiyue Deng
- Department of Neurobiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, P.R. China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Weiling Song
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Huan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
15
|
Paduszyński P, Aleksander-Konert E, Zajdel A, Wilczok A, Jelonek K, Witek A, Dzierżewicz Z. Changes in expression of cartilaginous genes during chondrogenesis of Wharton's jelly mesenchymal stem cells on three-dimensional biodegradable poly(L-lactide-co-glycolide) scaffolds. Cell Mol Biol Lett 2016; 21:14. [PMID: 28536617 PMCID: PMC5414664 DOI: 10.1186/s11658-016-0012-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/08/2016] [Indexed: 01/08/2023] Open
Abstract
Background In cartilage tissue regeneration, it is important to develop biodegradable scaffolds that provide a structural and logistic template for three-dimensional cultures of chondrocytes. In this study, we evaluated changes in expression of cartilaginous genes during in vitro chondrogenic differentiation of WJ-MSCs on PLGA scaffolds. Methods The biocompatibility of the PLGA material was investigated using WJ-MSCs by direct and indirect contact methods according to the ISO 10993–5 standard. PLGA scaffolds were fabricated by the solvent casting/salt-leaching technique. We analyzed expression of chondrogenic genes of WJ-MSCs after a 21-day culture. Results The results showed the biocompatibility of PLGA and confirmed the usefulness of PLGA as material for fabrication of 3D scaffolds that can be applied for WJ-MSC culture. The in vitro penetration and colonization of the scaffolds by WJ-MSCs were assessed by confocal microscopy. The increase in cell number demonstrated that scaffolds made of PLGA copolymers enabled WJ-MSC proliferation. The obtained data showed that as a result of chondrogenesis of WJ-MSCs on the PLGA scaffold the expression of the key markers collagen type II and aggrecan was increased. Conclusions The observed changes in transcriptional activity of cartilaginous genes suggest that the PLGA scaffolds may be applied for WJ-MSC differentiation. This primary study suggests that chondrogenic capacity of WJ-MSCs cultured on the PLGA scaffolds can be useful for cell therapy of cartilage.
Collapse
Affiliation(s)
- Piotr Paduszyński
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Ewelina Aleksander-Konert
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Alicja Zajdel
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Adam Wilczok
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Andrzej Witek
- Department of Gynecology and Obstetrics, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Zofia Dzierżewicz
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland.,Department of Health Care, Silesian Medical College, Katowice, Poland
| |
Collapse
|
16
|
James AW, Hindle P, Murray IR, West CC, Tawonsawatruk T, Shen J, Asatrian G, Zhang X, Nguyen V, Simpson AH, Ting K, Péault B, Soo C. Pericytes for the treatment of orthopedic conditions. Pharmacol Ther 2016; 171:93-103. [PMID: 27510330 DOI: 10.1016/j.pharmthera.2016.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 08/01/2016] [Indexed: 01/15/2023]
Abstract
Pericytes and other perivascular stem cells are of growing interest in orthopedics and tissue engineering. Long regarded as simple regulators of angiogenesis and blood pressure, pericytes are now recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Pericytes are typified by characteristic cell surface marker expression (including αSMA, CD146, PDGFRβ, NG2, RGS5, among others). Although alone no marker is absolutely specific for pericytes, collectively these markers appear to selectively identify an MSC-like pericyte. The purification of pericytes is most well described as a CD146+CD34-CD45- cell population. Pericytes and other perivascular stem cell populations have been applied in diverse orthopedic applications, including both ectopic and orthotopic models. Application of purified cells has sped calvarial repair, induced spine fusion, and prevented fibrous non-union in rodent models. Pericytes induce these effects via both direct and indirect mechanisms. In terms of their paracrine effects, pericytes are known to produce and secrete high levels of a number of growth and differentiation factors both in vitro and after transplantation. The following review will cover existing studies to date regarding pericyte application for bone and cartilage engineering. In addition, further questions in the field will be pondered, including the phenotypic and functional overlap between pericytes and culture-derived MSC, and the concept of pericytes as efficient producers of differentiation factors to speed tissue repair.
Collapse
Affiliation(s)
- Aaron W James
- School of Dentistry, University of California, Los Angeles, United States; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, United States; Orthopedic Hospital Research Center, University of California, Los Angeles, United States; Department of Pathology, Johns Hopkins University, Baltimore, MD, United States.
| | - Paul Hindle
- Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, United Kingdom
| | - Iain R Murray
- Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, United Kingdom; BHF Center for Vascular Regeneration & MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher C West
- BHF Center for Vascular Regeneration & MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom; Department of Plastic and Reconstructive Surgery, St. Johns Hospital, Livingston, United Kingdom
| | - Tulyapruek Tawonsawatruk
- Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, United Kingdom; BHF Center for Vascular Regeneration & MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom; Department of Orthopaedics, Ramathibodi Hospital, Madihol University, Thailand
| | - Jia Shen
- School of Dentistry, University of California, Los Angeles, United States
| | - Greg Asatrian
- School of Dentistry, University of California, Los Angeles, United States
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, United States
| | - Vi Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, United States
| | - A Hamish Simpson
- Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kang Ting
- School of Dentistry, University of California, Los Angeles, United States
| | - Bruno Péault
- Orthopedic Hospital Research Center, University of California, Los Angeles, United States; BHF Center for Vascular Regeneration & MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Chia Soo
- Orthopedic Hospital Research Center, University of California, Los Angeles, United States; Department of Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
17
|
Mesenchymal stem cells generate pericytes to promote tumor recurrence via vasculogenesis after stereotactic body radiation therapy. Cancer Lett 2016; 375:349-359. [DOI: 10.1016/j.canlet.2016.02.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/30/2016] [Accepted: 02/17/2016] [Indexed: 01/16/2023]
|
18
|
Yao Y, Shang J, Song W, Deng Q, Liu H, Zhou Y. Global profiling of the gene expression and alternative splicing events during hypoxia-regulated chondrogenic differentiation in human cartilage endplate-derived stem cells. Genomics 2016; 107:170-7. [PMID: 26996146 DOI: 10.1016/j.ygeno.2016.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/09/2016] [Accepted: 03/16/2016] [Indexed: 01/08/2023]
Abstract
The intervertebral disc (IVD) degeneration is initiated by cartilage endplate (CEP) degeneration and is characterised by reduced chondrification. Cartilage endplate-derived stem cells (CESCs) with chondrogenic differentiation abilities are responsible for the restoration of cartilage. CEP remains in an avascular and hypoxic microenvironment. In this study, we observed that the physiological hypoxia greatly promotes the chondrogenic differentiation of CESCs. This tissue specificity of the differentiation fate of CESCs in response to the hypoxic microenvironment was physiologically significant for the CEP to maintain the chondrification status. To investigate the mechanisms underlying the hypoxia-regulated chondrogenic differentiation of CESCs, we adopted a high-throughput scanning technology to detect the global profiling of gene expression and alternative splicing (AS) event changes during chondrogenic differentiation under hypoxia in CESCs compared to those induced under normoxia. An Affymetrix Human Transcriptome Array 2.0 was used to identify the differentially expressed genes (DEGs) and alternatively spliced genes (ASGs). After RT-PCR validation, GO and KEGG pathway analyses of both the DEGs and ASGs were performed. The enrichment of the GO functional terms and signalling pathways provided referential direction of the mechanism to study the gene expression and AS in the hypoxia-regulated chondrogenesis promotion, which could be helpful in understanding this physiological phenomenon, and it could also be instrumental in finding targets for CEP degeneration therapy.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jin Shang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Weilin Song
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Qiyue Deng
- Department of Neurobiology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Huan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
19
|
Reissis D, Tang QO, Cooper NC, Carasco CF, Gamie Z, Mantalaris A, Tsiridis E. Current clinical evidence for the use of mesenchymal stem cells in articular cartilage repair. Expert Opin Biol Ther 2016; 16:535-57. [PMID: 26798997 DOI: 10.1517/14712598.2016.1145651] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Articular cartilage is renowned for its poor intrinsic capacity for repair. Current treatments for osteoarthritis are limited in their ability to reliably restore the native articular cartilage structure and function. Mesenchymal stem cells (MSCs) present an attractive treatment option for articular cartilage repair, with a recent expansion of clinical trials investigating their use in patients. AREAS COVERED This paper provides a current overview of the clinical evidence on the use of MSCs in articular cartilage repair. EXPERT OPINION The article demonstrates robust clinical evidence that MSCs have significant potential for the regeneration of hyaline articular cartilage in patients. The majority of clinical trials to date have yielded significantly positive results with minimal adverse effects. However the clinical research is still in its infancy. The optimum MSC source, cell concentrations, implantation technique, scaffold, growth factors and rehabilitation protocol for clinical use are yet to be identified. A larger number of randomised control trials are required to objectively compare the clinical efficacy and long-term safety of the various techniques. As the clinical research continues to evolve and address these challenges, it is likely that MSCs may become integrated into routine clinical practice in the near future.
Collapse
Affiliation(s)
- Dimitris Reissis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Quen Oak Tang
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Nina Catherine Cooper
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Clare Francesca Carasco
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Zakareya Gamie
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Athanasios Mantalaris
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Eleftherios Tsiridis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK.,b Academic Orthopaedic Unit , Aristotle University Medical School , Thessaloniki , Greece
| |
Collapse
|
20
|
Foldager CB, Toh WS, Christensen BB, Lind M, Gomoll AH, Spector M. Collagen Type IV and Laminin Expressions during Cartilage Repair and in Late Clinically Failed Repair Tissues from Human Subjects. Cartilage 2016; 7:52-61. [PMID: 26958317 PMCID: PMC4749752 DOI: 10.1177/1947603515604022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To identify the collagen type IV (Col4) isoform in articular cartilage and to evaluate the expressions of Col4 and laminin in the pericellular matrix (PCM) in damaged cartilage and during cartilage repair. DESIGN The Col4 isoform was determined in chondrocytes isolated from 6 patients cultured up to 6 days and in 21% O2 or 1% O2, and the gene expression of Col4 α-chains was investigated. The distribution of Col4 and laminin in traumatically damaged cartilage (n = 7) and clinically failed cartilage repair (microfracture, TruFit, autologous chondrocyte implantation; n = 11) were investigated using immunohistochemistry. Normal human cartilage was used as control (n = 8). The distribution during clinical cartilage repair procedures was investigated in a minipig model with 6-month follow-up (untreated chondral, untreated osteochondral, microfracture, autologous chondrocyte implantation; n = 10). RESULTS The Col4 isoform in articular cartilage was characterized as α1α1α2, which is an isoform containing antiangiogenic domains in the NC1-terminals (arresten and canstatin). In normal cartilage, laminin and Col4 was exclusively found in the PCM. High amounts (>50%) of Col4 in the PCM significantly decreased in damaged cartilage (P = 0.004) and clinically failed repair tissue (P < 0.001). Laminin was only found with high expression (>50%) in 4/8 of the normal samples, which was not statistically significantly different from damaged cartilage (P = 0.15) or failed cartilage repair (P = 0.054). CONCLUSIONS Col4 in cartilage contain antiangiogenic domains and may play a role in the hypoxic environment in articular cartilage. Col4 and laminin was not found in the PCM of damaged and clinically failed repair.
Collapse
Affiliation(s)
- Casper Bindzus Foldager
- Department of Orthopedics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Tissue Engineering Labs, VA Boston Healthcare System, Boston, MA, USA
- Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Wei Seong Toh
- Department of Orthopedics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Tissue Engineering Labs, VA Boston Healthcare System, Boston, MA, USA
- Discipline of Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | | | - Martin Lind
- Sports Trauma Clinic, Department of Orthopaedics, Aarhus University Hospital, Aarhus, Denmark
| | - Andreas H. Gomoll
- Cartilage Repair Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Myron Spector
- Department of Orthopedics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Tissue Engineering Labs, VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
21
|
Lv FJ, Tuan RS, Cheung KMC, Leung VYL. Concise review: the surface markers and identity of human mesenchymal stem cells. Stem Cells 2015; 32:1408-19. [PMID: 24578244 DOI: 10.1002/stem.1681] [Citation(s) in RCA: 730] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 02/09/2014] [Indexed: 12/13/2022]
Abstract
The concept of mesenchymal stem cells (MSCs) is becoming increasingly obscure due to the recent findings of heterogeneous populations with different levels of stemness within MSCs isolated by traditional plastic adherence. MSCs were originally identified in bone marrow and later detected in many other tissues. Currently, no cloning based on single surface marker is capable of isolating cells that satisfy the minimal criteria of MSCs from various tissue environments. Markers that associate with the stemness of MSCs await to be elucidated. A number of candidate MSC surface markers or markers possibly related to their stemness have been brought forward so far, including Stro-1, SSEA-4, CD271, and CD146, yet there is a large difference in their expression in various sources of MSCs. The exact identity of MSCs in vivo is not yet clear, although reports have suggested they may have a fibroblastic or pericytic origin. In this review, we revisit the reported expression of surface molecules in MSCs from various sources, aiming to assess their potential as MSC markers and define the critical panel for future investigation. We also discuss the relationship of MSCs to fibroblasts and pericytes in an attempt to shed light on their identity in vivo.
Collapse
Affiliation(s)
- Feng-Juan Lv
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China; Stem Cell & Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong SAR, People's Republic of China; Center for Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | |
Collapse
|
22
|
Goude MC, McDevitt TC, Temenoff JS. Chondroitin sulfate microparticles modulate transforming growth factor-β1-induced chondrogenesis of human mesenchymal stem cell spheroids. Cells Tissues Organs 2014; 199:117-30. [PMID: 25413333 DOI: 10.1159/000365966] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2014] [Indexed: 11/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been previously explored as a part of cell-based therapies for the repair of damaged cartilage. Current MSC chondrogenic differentiation strategies employ large pellets; however, we have developed a technique to form small MSC aggregates (500-1,000 cells) that can reduce transport barriers while maintaining a multicellular structure analogous to cartilaginous condensations. The objective of this study was to examine the effects of incorporating chondroitin sulfate methacrylate (CSMA) microparticles (MPs) within small MSC spheroids cultured in the presence of transforming growth factor (TGF)-β1 on chondrogenesis. Spheroids with MPs induced earlier increases in collagen II and aggrecan gene expression (chondrogenic markers) than spheroids without MPs, although no large differences in immunostaining for these matrix molecules were observed by day 21 between these groups. Collagen I and X were also detected in the extracellular matrix (ECM) of all spheroids by immunostaining. Interestingly, histology revealed that CSMA MPs clustered together near the center of the MSC spheroids and induced circumferential alignment of cells and ECM around the material core. This study demonstrates the use of CSMA materials to further examine the effects of matrix molecules on MSC phenotype as well as potentially direct differentiation in a more spatially controlled manner that better mimics the architecture of specific musculoskeletal tissues.
Collapse
Affiliation(s)
- Melissa C Goude
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Ga., USA
| | | | | |
Collapse
|
23
|
Ni Y, Tang Z, Cao W, Lin H, Fan Y, Guo L, Zhang X. Tough and elastic hydrogel of hyaluronic acid and chondroitin sulfate as potential cell scaffold materials. Int J Biol Macromol 2014; 74:367-75. [PMID: 25445680 DOI: 10.1016/j.ijbiomac.2014.10.058] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 09/26/2014] [Accepted: 10/30/2014] [Indexed: 02/08/2023]
Abstract
Natural polysaccharides are extensively investigated as cell scaffold materials for cellular adhesion, proliferation, and differentiation due to their excellent biocompatibility, biodegradability, and biofunctions. However, their application is often severely limited by their mechanical behavior. In this study, a tough and elastic hydrogel scaffold was prepared with hyaluronic acid (HA) and chondroitin sulfate (CS). HA and CS were conjugated with tyramine (TA) and the degree of substitution (DS) was 10.7% and 11.3%, respectively, as calculated by (1)H NMR spectra. The hydrogel was prepared by mixing HA-TA and CS-TA in presence of H2O2 and HRP. The sectional morphology of hydrogels was observed by SEM, static and dynamic mechanical properties were analyzed by Shimadzu electromechanical testing machine and dynamic mechanical thermal analyzer Q800. All samples showed good ability to recover their appearances after deformation, the storage modulus (E') of hydrogels became higher as the testing frequency went up. Hydrogels also showed fatigue resistance to cyclic compression. Mesenchymal stem cells encapsulated in hydrogels showed good cell viability as detected by CLSM. This study suggests that the hydrogels have both good mechanical properties and biocompatibility, and may serve as model systems to explore mechanisms of deformation and energy dissipation or find some applications in tissue engineering.
Collapse
Affiliation(s)
- Yilu Ni
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China
| | - Zhurong Tang
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China
| | - Wanxu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China
| | - Hai Lin
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Wangjiang Road 29, Chengdu 610064, China.
| |
Collapse
|
24
|
Mazor M, Lespessailles E, Coursier R, Daniellou R, Best TM, Toumi H. Mesenchymal stem-cell potential in cartilage repair: an update. J Cell Mol Med 2014; 18:2340-50. [PMID: 25353372 PMCID: PMC4302639 DOI: 10.1111/jcmm.12378] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/27/2014] [Indexed: 01/05/2023] Open
Abstract
Articular cartilage damage and subsequent degeneration are a frequent occurrence in synovial joints. Treatment of these lesions is a challenge because this tissue is incapable of quality repair and/or regeneration to its native state. Non-operative treatments endeavour to control symptoms and include anti-inflammatory medications, viscosupplementation, bracing, orthotics and activity modification. Classical surgical techniques for articular cartilage lesions are frequently insufficient in restoring normal anatomy and function and in many cases, it has not been possible to achieve the desired results. Consequently, researchers and clinicians are focusing on alternative methods for cartilage preservation and repair. Recently, cell-based therapy has become a key focus of tissue engineering research to achieve functional replacement of articular cartilage. The present manuscript is a brief review of stem cells and their potential in the treatment of early OA (i.e. articular cartilage pathology) and recent progress in the field.
Collapse
Affiliation(s)
- M Mazor
- IPROS, CHRO, EA4708 Orleans University, Orleans, France
| | | | | | | | | | | |
Collapse
|
25
|
Boyette LB, Creasey OA, Guzik L, Lozito T, Tuan RS. Human bone marrow-derived mesenchymal stem cells display enhanced clonogenicity but impaired differentiation with hypoxic preconditioning. Stem Cells Transl Med 2014; 3:241-54. [PMID: 24436440 DOI: 10.5966/sctm.2013-0079] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells are promising candidate cells for regenerative applications because they possess high proliferative capacity and the potential to differentiate into other cell types. Mesenchymal stem cells (MSCs) are easily sourced but do not retain their proliferative and multilineage differentiative capabilities after prolonged ex vivo propagation. We investigated the use of hypoxia as a preconditioning agent and in differentiating cultures to enhance MSC function. Culture in 5% ambient O(2) consistently enhanced clonogenic potential of primary MSCs from all donors tested. We determined that enhanced clonogenicity was attributable to increased proliferation, increased vascular endothelial growth factor secretion, and increased matrix turnover. Hypoxia did not impact the incidence of cell death. Application of hypoxia to osteogenic cultures resulted in enhanced total mineral deposition, although this effect was detected only in MSCs preconditioned in normoxic conditions. Osteogenesis-associated genes were upregulated in hypoxia, and alkaline phosphatase activity was enhanced. Adipogenic differentiation was inhibited by exposure to hypoxia during differentiation. Chondrogenesis in three-dimensional pellet cultures was inhibited by preconditioning with hypoxia. However, in cultures expanded under normoxia, hypoxia applied during subsequent pellet culture enhanced chondrogenesis. Whereas hypoxic preconditioning appears to be an excellent way to expand a highly clonogenic progenitor pool, our findings suggest that it may blunt the differentiation potential of MSCs, compromising their utility for regenerative tissue engineering. Exposure to hypoxia during differentiation (post-normoxic expansion), however, appears to result in a greater quantity of functional osteoblasts and chondrocytes and ultimately a larger quantity of high-quality differentiated tissue.
Collapse
Affiliation(s)
- Lisa B Boyette
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, McGowan Institute for Regenerative Medicine, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
26
|
Brown PT, Handorf AM, Jeon WB, Li WJ. Stem cell-based tissue engineering approaches for musculoskeletal regeneration. Curr Pharm Des 2013; 19:3429-45. [PMID: 23432679 DOI: 10.2174/13816128113199990350] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/10/2013] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine and tissue engineering is an ever evolving field that holds promise in treating numerous musculoskeletal diseases and injuries. An important impetus in the development of the field was the discovery and implementation of stem cells. The utilization of mesenchymal stem cells, and later embryonic and induced pluripotent stem cells, opens new arenas for tissue engineering and presents the potential of developing stem cell-based therapies for disease treatment. Multipotent and pluripotent stem cells can produce various lineage tissues, and allow for derivation of a tissue that may be comprised of multiple cell types. As the field grows, the combination of biomaterial scaffolds and bioreactors provides methods to create an environment for stem cells that better represent their microenvironment for new tissue formation. As technologies for the fabrication of biomaterial scaffolds advance, the ability of scaffolds to modulate stem cell behavior advances as well. The composition of scaffolds could be of natural or synthetic materials and could be tailored to enhance cell self-renewal and/or direct cell fates. In addition to biomaterial scaffolds, studies of tissue development and cellular microenvironments have determined other factors, such as growth factors and oxygen tension, that are crucial to the regulation of stem cell activity. The overarching goal of stem cell-based tissue engineering research is to precisely control differentiation of stem cells in culture. In this article, we review current developments in tissue engineering, focusing on several stem cell sources, induction factors including growth factors, oxygen tension, biomaterials, and mechanical stimulation, and the internal and external regulatory mechanisms that govern proliferation and differentiation.
Collapse
Affiliation(s)
- Patrick T Brown
- Wisconsin Institutes of Medical Research, 1111 Highland Ave., Madison, WI 53705, USA
| | | | | | | |
Collapse
|
27
|
|
28
|
de Vries-van Melle ML, Narcisi R, Kops N, Koevoet WJLM, Bos PK, Murphy JM, Verhaar JAN, van der Kraan PM, van Osch GJVM. Chondrogenesis of mesenchymal stem cells in an osteochondral environment is mediated by the subchondral bone. Tissue Eng Part A 2013; 20:23-33. [PMID: 23980750 DOI: 10.1089/ten.tea.2013.0080] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In articular cartilage repair, cells that will be responsible for the formation of repair tissue are often exposed to an osteochondral environment. To study cartilage repair mechanisms in vitro, we have recently developed a bovine osteochondral biopsy culture model in which cartilage defects can be simulated reproducibly. Using this model, we now aimed at studying the chondrogenic potential of human bone marrow-derived mesenchymal stem cells (hBMSCs) in an osteochondral environment. In contrast to standard in vitro chondrogenesis, it was found that supplementing transforming growth factor beta (TGFβ) to culture medium was not required to induce chondrogenesis of hBMSCs in an osteochondral environment. hBMSC culture in defects created in osteochondral biopsies or in bone-only biopsies resulted in comparable levels of cartilage-related gene expression, whereas culture in cartilage-only biopsies did not induce chondrogenesis. Subcutaneous implantation in nude mice of osteochondral biopsies containing hBMSCs in osteochondral defects resulted in the formation of more cartilaginous tissue than hBMSCs in chondral defects. The subchondral bone secreted TGFβ; however, the observed results could not be attributed to TGFβ, as either capturing TGFβ with an antibody or blocking the canonical TGFβ signaling pathway did not result in significant changes in cartilage-related gene expression of hBMSCs in the osteochondral culture model. Inhibition of BMP signaling did not prevent chondrogenesis. In conclusion, we demonstrate that chondrogenesis of hBMSCs is induced by factors secreted from the bone. We have strong indications that this is not solely mediated by members of the TGFβ family but other, yet unknown, factors originating from the subchondral bone appeared to play a key role.
Collapse
|
29
|
Garza-Veloz I, Romero-Diaz VJ, Martinez-Fierro ML, Marino-Martinez IA, Gonzalez-Rodriguez M, Martinez-Rodriguez HG, Espinoza-Juarez MA, Bernal-Garza DA, Ortiz-Lopez R, Rojas-Martinez A. Analyses of chondrogenic induction of adipose mesenchymal stem cells by combined co-stimulation mediated by adenoviral gene transfer. Arthritis Res Ther 2013; 15:R80. [PMID: 23899094 PMCID: PMC3978573 DOI: 10.1186/ar4260] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/30/2013] [Indexed: 12/20/2022] Open
Abstract
Introduction Adipose-derived stem cells (ASCs) have the potential to differentiate into cartilage under stimulation with some reported growth and transcriptional factors, which may constitute an alternative for cartilage replacement approaches. In this study, we analyzed the in vitro chondrogenesis of ASCs transduced with adenoviral vectors encoding insulin-like growth factor-1 (IGF-1), transforming growth factor beta-1 (TGF-β1), fibroblast growth factor-2 (FGF-2), and sex-determining region Y-box 9 (SOX9) either alone or in combinations. Methods Aggregate cultures of characterized ovine ASCs were transduced with 100 multiplicity of infections of Ad.IGF-1, Ad.TGF-β1, Ad.FGF-2, and Ad.SOX9 alone or in combination. These were harvested at various time points for detection of cartilage-specific genes expression by quantitative real-time PCR or after 14 and 28 days for histologic and biochemical analyses detecting proteoglycans, collagens (II, I and X), and total sulfated glycosaminoglycan and collagen content, respectively. Results Expression analyses showed that co-expression of IGF-1 and FGF-2 resulted in higher significant expression levels of aggrecan, biglycan, cartilage matrix, proteoglycan, and collagen II (all P ≤0.001 at 28 days). Aggregates co-transduced with Ad.IGF-1/Ad.FGF-2 showed a selective expression of proteoglycans and collagen II, with limited expression of collagens I and × demonstrated by histological analyses, and had significantly greater glycosaminoglycan and collagen production than the positive control (P ≤0.001). Western blot analyses for this combination also demonstrated increased expression of collagen II, while expression of collagens I and × was undetectable and limited, respectively. Conclusion Combined overexpression of IGF-1/FGF-2 within ASCs enhances their chondrogenic differentiation inducing the expression of chondrogenic markers, suggesting that this combination is more beneficial than the other factors tested for the development of cell-based therapies for cartilage repair.
Collapse
|
30
|
Andreeva ER, Buravkova LB. Paracrine activity of multipotent mesenchymal stromal cells and its modulation in hypoxia. ACTA ACUST UNITED AC 2013. [DOI: 10.1134/s0362119713030043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Preitschopf A, Zwickl H, Li K, Lubec G, Joo G, Rosner M, Hengstschläger M, Mikula M. Chondrogenic differentiation of amniotic fluid stem cells and their potential for regenerative therapy. Stem Cell Rev Rep 2013; 8:1267-74. [PMID: 22869300 DOI: 10.1007/s12015-012-9405-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic articular cartilage defects are the most common disabling conditions of humans in the western world. The incidence for cartilage defects is increasing with age and the most prominent risk factors are overweight and sports associated overloading. Damage of articular cartilage frequently leads to osteoarthritis due to the aneural and avascular nature of articular cartilage, which impairs regeneration and repair. Hence, patients affected by cartilage defects will benefit from a cell-based transplantation strategy. Autologous chondrocytes, mesenchymal stem cells and embryonic stem cells are suitable donor cells for regeneration approaches and most recently the discovery of amniotic fluid stem cells has opened a plethora of new therapeutic options. It is the aim of this review to summarize recent advances in the use of amniotic fluid stem cells as novel cell sources for the treatment of articular cartilage defects. Molecular aspects of articular cartilage formation as well as degeneration are summarized and the role of growth factor triggered signaling pathways, scaffolds, hypoxia and autophagy during the process of chondrogenic differentiation are discussed.
Collapse
Affiliation(s)
- Andrea Preitschopf
- Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Miyagi S, Tensho K, Wakitani S, Takagi M. Construction of an osteochondral-like tissue graft combining β-tricalcium phosphate block and scaffold-free mesenchymal stem cell sheet. J Orthop Sci 2013; 18:471-7. [PMID: 23471715 DOI: 10.1007/s00776-013-0368-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 02/12/2013] [Indexed: 02/09/2023]
Abstract
BACKGROUND Aiming to construct an osteochondral-like structure, the combination of a β-tricalcium phosphate (βTCP) block with a scaffold-free sheet formed using mesenchymal stem cells (MSCs) was investigated. METHODS Human bone marrow MSCs in a cell culture insert that was set in a 24-well plate were cultivated using a chondrogenic medium containing dexamethasone, IGF-1, and TGFβ3 for 3 weeks during which a cylindrical βTCP block was put on the sheet at day 1, and the cell sheet construct was harvested. In other experiments, at day 14, the construct was put on a cell sheet that was prepared the day before and cultivated for 3 weeks. RESULTS The addition of a βTCP block resulted in a combined osteochondral-like construct and comparable staining intensity by Alcian blue, while the expression levels of the aggrecan and type II collagen genes decreased a little. During the culture with the βTCP block, the expression levels of the aggrecan gene increased monotonically. The increase in the inoculum cell number from 1.86 to 3.72 × 10(6) cells resulted in marked increases in the thickness of cell sheet parts in the βTCP block and expression levels of the aggrecan and type II collagen genes, while the thickness of cell sheet parts on the βTCP block scarcely changed. On the other hand, the addition of a cell sheet that was prepared a day before to the construct at day 14 resulted in the marked increase in thickness of the cell sheet part on the βTCP block, while the thickness of that in the βTCP block did not increase. CONCLUSION A combined osteochondral-like structure was produced by putting a βTCP block on the sheet of MSC. The thickness of the cell sheet parts in and on the βTCP block could be increased by the increase in inoculum cell number and by providing an additional cell sheet, respectively.
Collapse
Affiliation(s)
- Shigeharu Miyagi
- Division of Biotechnology and Macromolecular Chemistry, Graduate School of Engineering, Hokkaido University, N13W8 Kita-ku, Sapporo, Hokkaido, 060-8628, Japan
| | | | | | | |
Collapse
|
33
|
Gornostaeva AN, Andreeva ER, Buravkova LB. Human MMSC immunosuppressive activity at low oxygen tension: Direct cell-to-cell contacts and paracrine regulation. ACTA ACUST UNITED AC 2013. [DOI: 10.1134/s0362119713020059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
34
|
Surface markers for chondrogenic determination: a highlight of synovium-derived stem cells. Cells 2012; 1:1107-20. [PMID: 24710545 PMCID: PMC3901147 DOI: 10.3390/cells1041107] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 10/31/2012] [Accepted: 11/12/2012] [Indexed: 02/06/2023] Open
Abstract
Cartilage tissue engineering is a promising field in regenerative medicine that can provide substantial relief to people suffering from degenerative cartilage disease. Current research shows the greatest chondrogenic potential for healthy articular cartilage growth with minimal hypertrophic differentiation to be from mesenchymal stem cells (MSCs) of synovial origin. These stem cells have the capacity for differentiation into multiple cell lineages related to mesenchymal tissue; however, evidence exists for cell surface markers that specify a greater potential for chondrogenesis than other differentiation fates. This review will examine relevant literature to summarize the chondrogenic differentiation capacities of tested synovium-derived stem cell (SDSC) surface markers, along with a discussion about various other markers that may hold potential, yet require further investigation. With this information, a potential clinical benefit exists to develop a screening system for SDSCs that will produce the healthiest articular cartilage possible.
Collapse
|
35
|
Beane OS, Darling EM. Isolation, characterization, and differentiation of stem cells for cartilage regeneration. Ann Biomed Eng 2012; 40:2079-97. [PMID: 22907257 DOI: 10.1007/s10439-012-0639-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/08/2012] [Indexed: 12/27/2022]
Abstract
The goal of tissue engineering is to create a functional replacement for tissues damaged by injury or disease. In many cases, impaired tissues cannot provide viable cells, leading to the investigation of stem cells as a possible alternative. Cartilage, in particular, may benefit from the use of stem cells since the tissue has low cellularity and cannot effectively repair itself. To address this need, researchers are investigating the chondrogenic capabilities of several multipotent stem cell sources, including adult and extra-embryonic mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). Comparative studies indicate that each cell type has advantages and disadvantages, and while direct comparisons are difficult to make, published data suggest some sources may be more promising for cartilage regeneration than others. In this review, we identify current approaches for isolating and chondrogenically differentiating MSCs from bone marrow, fat, synovium, muscle, and peripheral blood, as well as cells from extra-embryonic tissues, ESCs, and iPSCs. Additionally, we assess chondrogenic induction with growth factors, identifying standard cocktails used for each stem cell type. Cell-only (pellet) and scaffold-based studies are also included, as is a discussion of in vivo results.
Collapse
Affiliation(s)
- Olivia S Beane
- Center for Biomedical Engineering, Brown University, Providence, RI, USA
| | | |
Collapse
|
36
|
Buravkova LB, Andreeva ER, Grigoriev AI. The impact of oxygen in physiological regulation of human multipotent mesenchymal cell functions. ACTA ACUST UNITED AC 2012. [DOI: 10.1134/s0362119712040032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
37
|
Hypoxia mediated isolation and expansion enhances the chondrogenic capacity of bone marrow mesenchymal stromal cells. Stem Cell Res Ther 2012; 3:9. [PMID: 22385573 PMCID: PMC3392769 DOI: 10.1186/scrt100] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/18/2011] [Accepted: 03/02/2012] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The capacity of bone marrow mesenchymal stromal cells (BMSCs) to be induced into chondrocytes has drawn much attention for cell-based cartilage repair. BMSCs represent a small proportion of cells of the bone marrow stromal compartment and, thus, culture expansion is a necessity for therapeutic use. However, there is no consensus on how BMSCs should be isolated nor expanded to maximize their chondrogenic potential. During embryonic development pluripotent stem cells differentiate into chondrocytes and form cartilage in a hypoxic microenvironment. METHODS Freshly harvested human BMSCs were isolated and expanded from the aspirates of six donors, under either hypoxic conditions (3% O2) or normoxic conditions (21% O2). A colony-forming unit fibroblastic (Cfu-f) assay was used to determine the number of cell colonies developed from each donor. BMSCs at passage 2 (P2) were characterized by flow cytometry for the phenotypic expression of cell surface markers on mesenchymal stem cells. BMSCs at P2 were subsequently cultured in vitro as three-dimensional cell pellets in a defined serum-free chondrogenic medium under normoxic and hypoxic conditions. Chondrogenic differentiation of the BMSCs was characterized by biochemical and histological methods and by quantitative gene-expression analysis. RESULTS After 14 days of culture, the number of BMSC colonies developed under hypoxia was generally higher (8% to 38% depending on donor) than under normoxia. BMSCs were positive for the cell surface markers CD13, CD29, CD44, CD73, CD90, CD105 and CD151, and negative for CD34. Regardless of the oxygen tension during pellet culture, hypoxia-expanded BMSC pellets underwent a more robust chondrogenesis than normoxia-expanded BMSC pellets after three weeks of culture, as judged by increased glycosaminoglycan synthesis and Safranin O staining, along with increased mRNA expression of aggrecan, collagen II and Sox9. Hypoxic conditions enhanced the mRNA expression of hypoxia inducible factor-2 alpha (HIF-2α) but suppressed the mRNA expression of collagen X in BMSC pellet cultures regardless of the oxygen tension during BMSC isolation and propagation. CONCLUSIONS Taken together, our data demonstrate that isolation and expansion of BMSCs under hypoxic conditions augments the chondrogenic potential of BMSCs. This suggests that hypoxia-mediated isolation and expansion of BMSCs may improve clinical applications of BMSCs for cartilage repair.
Collapse
|
38
|
Freyria AM, Mallein-Gerin F. Chondrocytes or adult stem cells for cartilage repair: the indisputable role of growth factors. Injury 2012; 43:259-65. [PMID: 21696723 DOI: 10.1016/j.injury.2011.05.035] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 05/25/2011] [Indexed: 02/02/2023]
Abstract
Articular cartilage is easily injured but difficult to repair and cell therapies are proposed as tools to regenerate the defects in the tissue. Both differentiated chondrocytes and adult mesenchymal stem cells (MSCs) are regarded as cells potentially able to restore a functional cartilage. However, it is a complex process from the cell level to the tissue end product, during which growth factors play important roles from cell proliferation, extracellular matrix synthesis, maintenance of the phenotype to induction of MSCs towards chondrogenesis. Members of the TGF-β superfamily, are especially important in fulfilling these roles. Depending on the cell type chosen to restore cartilage, the effect of growth factors will vary. In this review, the roles of these factors in the maintenance of the chondrocyte phenotype are discussed and compared with those of factors involved in the repair of cartilage defects, using chondrocytes or adult mesenchymal stem cells.
Collapse
Affiliation(s)
- Anne-Marie Freyria
- Cartilage Biology and Engineering Group, IBCP, Université Lyon 1, Univ Lyon, CNRS FRE 3310, IFR128, France.
| | | |
Collapse
|
39
|
Egli RJ, Wernike E, Grad S, Luginbühl R. Physiological cartilage tissue engineering effect of oxygen and biomechanics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 289:37-87. [PMID: 21749898 DOI: 10.1016/b978-0-12-386039-2.00002-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vitro engineering of cartilaginous tissues has been studied for many years, and tissue-engineered constructs are sought to be used clinically for treating articular cartilage defects. Even though there is a plethora of studies and data available, no breakthroughs have been achieved yet that allow for implanting in vivo cultured articular cartilaginous tissues in patients. A review of contributions to cartilage tissue engineering over the past decades emphasizes that most of the studies were performed under environmental conditions neglecting the physiological situation. This is specifically pronounced in the use of bioreactor systems which neither allow for application of near physiomechanical stimulations nor for controlling a hypoxic environment as it is experienced in synovial joints. It is suspected that the negligence of these important parameters has slowed down progress and prevented major breakthroughs in the field. This review focuses on the main aspects of cartilage tissue engineering with emphasis on the relation and understanding of employing physiological conditions.
Collapse
|
40
|
Khan WS, Adesida AB, Tew SR, Longo UG, Hardingham TE. Fat pad-derived mesenchymal stem cells as a potential source for cell-based adipose tissue repair strategies. Cell Prolif 2012; 45:111-20. [PMID: 22260253 DOI: 10.1111/j.1365-2184.2011.00804.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells are able to undergo adipogenic differentiation and present a possible alternative cell source for regeneration and replacement of adipose tissue. The human infrapatellar fat pad is a promising source of mesenchymal stem cells with many source advantages over from bone marrow. It is important to determine whether a potential mesenchymal stem-cell exhibits tri-lineage differentiation potential and is able to maintain its proliferation potential and cell-surface characterization on expansion in tissue culture. We have previously shown that mesenchymal stem cells derived from the fat pad can undergo chondrogenic and osteogenic differentiation, and we characterized these cells at early passage. In the study described here, proliferation potential and characterization of fat pad-derived mesenchymal stem cells were assessed at higher passages, and cells were allowed to undergo adipogenic differentiation. MATERIALS AND METHODS Infrapatellar fat pad tissue was obtained from six patients undergoing total knee replacement. Cells isolated were expanded to passage 18 and proliferation rates were measured. Passage 10 and 18 cells were characterized for cell-surface epitopes using a range of markers. Passage 2 cells were allowed to undergo differentiation in adipogenic medium. RESULTS The cells maintained their population doubling rates up to passage 18. Cells at passage 10 and passage 18 had cell-surface epitope expression similar to other mesenchymal stem cells previously described. By staining it was revealed that they highly expressed CD13, CD29, CD44, CD90 and CD105, and did not express CD34 or CD56, they were also negative for LNGFR and STRO1. 3G5 positive cells were noted in cells from both passages. These fat pad-derived cells had adipogenic differentiation when assessed using gene expression for peroxisome proliferator-activated receptor γ2 and lipoprotein lipase, and oil red O staining. DISCUSSION These results indicate that the cells maintained their proliferation rate, and continued expressing mesenchymal stem-cell markers and pericyte marker 3G5 at late passages. These results also show that the cells were capable of adipogenic differentiation and thus could be a promising source for regeneration and replacement of adipose tissue in reconstructive surgery.
Collapse
Affiliation(s)
- W S Khan
- United Kingdom Centre for Tissue Engineering, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
41
|
Yeung TY, Seeberger KL, Kin T, Adesida A, Jomha N, Shapiro AMJ, Korbutt GS. Human mesenchymal stem cells protect human islets from pro-inflammatory cytokines. PLoS One 2012; 7:e38189. [PMID: 22666480 PMCID: PMC3364233 DOI: 10.1371/journal.pone.0038189] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/01/2012] [Indexed: 02/07/2023] Open
Abstract
Transplantation of human islets is an attractive alternative to daily insulin injections for patients with type 1 diabetes. However, the majority of islet recipients lose graft function within five years. Inflammation is a primary contributor to graft loss, and inhibiting pro-inflammatory cytokine activity can reverse inflammation mediated dysfunction of islet grafts. As mesenchymal stem cells (MSCs) possess numerous immunoregulatory properties, we hypothesized that MSCs could protect human islets from pro-inflammatory cytokines. Five hundred human islets were co-cultured with 0.5 or 1.0 × 10(6) human MSCs derived from bone marrow or pancreas for 24 hours followed by 48 hour exposure to interferon-γ, tumor necrosis factor-α and interleukin 1β. Controls include islets cultured alone (± cytokines) and with human dermal fibroblasts (± cytokines). For all conditions, glucose stimulated insulin secretion (GSIS), total islet cellular insulin content, islet β cell apoptosis, and potential cytoprotective factors secreted in the culture media were determined. Cytokine exposure disrupted human islet GSIS based on stimulation index and percentage insulin secretion. Conversely, culture with 1.0 × 10(6) bMSCs preserved GSIS from cytokine treated islets. Protective effects were not observed with fibroblasts, indicating that preservation of human islet GSIS after exposure to pro-inflammatory cytokines is MSC dependent. Islet β cell apoptosis was observed in the presence of cytokines; however, culture of bMSCs with islets prevented β cell apoptosis after cytokine treatment. Hepatocyte growth factor (HGF) as well as matrix metalloproteinases 2 and 9 were also identified as putative secreted cytoprotective factors; however, other secreted factors likely play a role in protection. This study, therefore, demonstrates that MSCs may be beneficial for islet engraftment by promoting cell survival and reduced inflammation.
Collapse
Affiliation(s)
- Telford Y. Yeung
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Karen L. Seeberger
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Tatsuya Kin
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Adetola Adesida
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Nadr Jomha
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - A. M. James Shapiro
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory S. Korbutt
- Department of Surgery, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, 5-002 Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
42
|
The potential of stem cells in the treatment of skeletal muscle injury and disease. Stem Cells Int 2011; 2012:282348. [PMID: 22220178 PMCID: PMC3246792 DOI: 10.1155/2012/282348] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 09/18/2011] [Indexed: 01/05/2023] Open
Abstract
Tissue engineering is a pioneering field with huge advances in recent times. These advances are not only in the understanding of how cells can be manipulated but also in potential clinical applications. Thus, tissue engineering, when applied to skeletal muscle cells, is an area of huge prospective benefit to patients with muscle disease/damage. This could include damage to muscle from trauma and include genetic abnormalities, for example, muscular dystrophies. Much of this research thus far has been focused on satellite cells, however, mesenchymal stem cells have more recently come to the fore. In particular, results of trials and further research into their use in heart failure, stress incontinence, and muscular dystrophies are eagerly awaited. Although no doubt, stem cells will have much to offer in the future, the results of further research still limit their use.
Collapse
|
43
|
Mahmoudifar N, Doran PM. Chondrogenesis and cartilage tissue engineering: the longer road to technology development. Trends Biotechnol 2011; 30:166-76. [PMID: 22071143 DOI: 10.1016/j.tibtech.2011.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 09/20/2011] [Accepted: 09/20/2011] [Indexed: 12/31/2022]
Abstract
Joint injury and disease are painful and debilitating conditions affecting a substantial proportion of the population. The idea that damaged cartilage in articulating joints might be replaced seamlessly with tissue-engineered cartilage is of obvious commercial interest because the market for such treatments is large. Recently, a wealth of new information about the complex biology of chondrogenesis and cartilage has emerged from stem cell research, including increasing evidence of the role of physical stimuli in directing differentiation. The challenge for the next generation of tissue engineers is to identify the key elements in this new body of knowledge that can be applied to overcome current limitations affecting cartilage synthesis in vitro. Here we review the status of cartilage tissue engineering and examine the contribution of stem cell research to technology development for cartilage production.
Collapse
Affiliation(s)
- Nastaran Mahmoudifar
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | | |
Collapse
|
44
|
Huang S, Leung V, Peng S, Li L, Lu FJ, Wang T, Lu W, Cheung KMC, Zhou G. Developmental definition of MSCs: new insights into pending questions. Cell Reprogram 2011; 13:465-72. [PMID: 21919705 DOI: 10.1089/cell.2011.0045] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a rare heterogeneous population of multipotent cells that can be isolated from many different adult and fetal tissues. They exhibit the capacity to give rise to cells of multiple lineages and are defined by their phenotype and functional properties, such as spindle-shaped morphology, adherence to plastic, immune response modulation capacity, and multilineage differentiation potential. Accordingly, MSCs have a wide range of promising applications in the treatment of autoimmune diseases, tissue repair, and regeneration. Recent studies have shed some light on the exact identity and native distribution of MSCs, whereas controversial results are still being reported, indicating the need for further review on their definition and origin. In this article, we summarize the important progress and describe some of our own relevant work on the developmental definition of MSCs.
Collapse
Affiliation(s)
- Shishu Huang
- Department of Orthopaedics and Traumatology, the University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Punwar S, Khan WS. Mesenchymal stem cells and articular cartilage repair: clinical studies and future direction. Open Orthop J 2011. [PMID: 21886696 DOI: 10.2174/187432500110501029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cartilage is frequently injured but shows little capacity for repair. Current treatment options include the use of procedures that stimulate repair through the stimulation of subchondral bone marrow and result in the formation of fibrocartilage. There is considerable interest in the use of cell-based treatment strategies and there are limited studies describing the use of mesenchymal stem cells for cartilage repair with promising early results. This paper reviews the current treatment strategies for articular cartilage, describes use of mesenchymal stem cells for articular cartilage repair along with the results of clinical studies, and describes the future direction that these strategies are likely to take.
Collapse
Affiliation(s)
- Shahid Punwar
- Department of Trauma and Orthopaedics, Frenchay Hospital, North Bristol NHS Trust, Bristol, BS16 1LE, UK
| | | |
Collapse
|
46
|
Punwar S, Khan WS. Mesenchymal stem cells and articular cartilage repair: clinical studies and future direction. Open Orthop J 2011; 5 Suppl 2:296-301. [PMID: 21886696 PMCID: PMC3149861 DOI: 10.2174/1874325001105010296] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 02/24/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022] Open
Abstract
Cartilage is frequently injured but shows little capacity for repair. Current treatment options include the use of procedures that stimulate repair through the stimulation of subchondral bone marrow and result in the formation of fibrocartilage. There is considerable interest in the use of cell-based treatment strategies and there are limited studies describing the use of mesenchymal stem cells for cartilage repair with promising early results. This paper reviews the current treatment strategies for articular cartilage, describes use of mesenchymal stem cells for articular cartilage repair along with the results of clinical studies, and describes the future direction that these strategies are likely to take.
Collapse
Affiliation(s)
- Shahid Punwar
- Department of Trauma and Orthopaedics, Frenchay Hospital, North Bristol NHS Trust, Bristol, BS16 1LE, UK
| | | |
Collapse
|
47
|
|
48
|
Circadian rhythm and cartilage extracellular matrix genes in osseointegration: a genome-wide screening of implant failure by vitamin D deficiency. PLoS One 2011; 6:e15848. [PMID: 21264318 PMCID: PMC3019224 DOI: 10.1371/journal.pone.0015848] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 11/25/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Successful dental and orthopedic implants require the establishment of an intimate association with bone tissue; however, the mechanistic explanation of how biological systems accomplish osseointegration is still incomplete. We sought to identify critical gene networks involved in osseointegration by exploring the implant failure model under vitamin D deficiency. METHODOLOGY Adult male Sprague-Dawley rats were exposed to control or vitamin D-deficient diet prior to the osteotomy surgery in the femur bone and the placement of T-shaped Ti4Al6V implant. Two weeks after the osteotomy and implant placement, tissue formed at the osteotomy site or in the hollow chamber of T-shaped implant was harvested and total RNA was evaluated by whole genome microarray analyses. PRINCIPAL FINDINGS Two-way ANOVA of microarray data identified 103 genes that were significantly (>2 fold) modulated by the implant placement and vitamin D deficiency. Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses assigned the highest z-score to the circadian rhythm pathway including neuronal PAS domain 2 (NPAS2), and period homolog 2 (Per2). NPAS2 and Aryl hydrocarbon receptor nuclear translocator-like (ARNTL/Bmal 1) were upregulated around implant and diminished by vitamin D deficiency, whereas the expression pattern of Per2 was complementary. Hierarchical cluster analysis further revealed that NPAS2 was in a group predominantly composed of cartilage extracellular matrix (ECM) genes. Whereas the expression of bone ECM genes around implant was not significantly affected by vitamin D deficiency, cartilage ECM genes were modulated by the presence of the implant and vitamin D status. In a proof-of-concept in vitro study, the expression of cartilage type II and X collagens was found upregulated when mouse mesenchymal stem cells were cultured on implant disk with 1,25D supplementation. CONCLUSIONS This study suggests that the circadian rhythm system and cartilage extracellular matrix may be involved in the establishment of osseointegration under vitamin D regulation.
Collapse
|
49
|
Si YL, Zhao YL, Hao HJ, Fu XB, Han WD. MSCs: Biological characteristics, clinical applications and their outstanding concerns. Ageing Res Rev 2011; 10:93-103. [PMID: 20727988 DOI: 10.1016/j.arr.2010.08.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/29/2010] [Accepted: 08/04/2010] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) are multi-potent adult stem cells harboring multi-lineage differentiation potential and immunosuppressive properties that make MSCs an ideal candidate cell type for immunomodulation and regenerative medicine. Currently, MSC-related researches and clinical trials have evoked exciting promise in a variety of disorders and tissue regeneration. However, it must be recognized that several critical potential problems have also emerged from current clinical trials, for example: (1) the indefinite association between the phenotypic characteristics and the biological functions of MSCs; (2) the lack of clinical data to support the long-term safety of MSCs; (3) the need for further clarification of multiple mechanisms of MSC transplant actions in vivo; and (4) the lack of comparability of MSC transplant efficacy. Therefore, MSC-based therapies could not yet be considered a routine treatment in the clinic. Based on these, we proposed that large-scale and multi-center clinical trials of MSC-based therapies should be initiated under strict supervision. These interventions might help to establish a new clinical paradigm to turn MSC transplantation into a routine therapy for at least some diseases in the near future.
Collapse
Affiliation(s)
- Yi-Ling Si
- Institute of Basic Medicine Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | | | | | | | | |
Collapse
|
50
|
Khan WS, Johnson DS, Hardingham TE. The potential of stem cells in the treatment of knee cartilage defects. Knee 2010; 17:369-74. [PMID: 20051319 DOI: 10.1016/j.knee.2009.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 12/06/2009] [Accepted: 12/07/2009] [Indexed: 02/02/2023]
Abstract
Cartilage is frequently damaged but only shows a limited capacity for repair. There are a number of treatment strategies currently available for the repair of articular cartilage defects including abrasion chondroplasty, subchondral drilling, microfracture and mosaicplasty but these show variable results. For the younger patients, there is great interest in the potential of cell-based strategies to provide a biological replacement of damaged cartilage using autologous chondrocytes. The results of clinical studies using these cell-based techniques do not conclusively show improvement over conventional techniques. These techniques also do not consistently result in the formation of the desired hyaline cartilage rather than fibrocartilage. Mesenchymal stem cells present a promising cell source for cartilage repair. Mesenchymal stem cells have been isolated from a number of adult tissues including the bone marrow and the synovial fat pad. These cells have the ability to proliferate in culture and differentiate down different pathways including the chondrogenic pathway. In the first instance, differentiated stem cells can be used for the repair of localised cartilage defects by producing hyaline cartilage. In the future, this strategy has the potential to be extended to treat more generalised cartilage defects, especially as the cell source is not a limiting factor. The use of cell-based therapies also allows the versatility of using scaffolds and growth factors, with recombinant proteins or gene therapy. A number of challenges however still need to be overcome including further work on identifying the optimal source of stem cells, along with refining the conditions that enhance expansion and chondrogenesis.
Collapse
Affiliation(s)
- Wasim S Khan
- UK Centre for Tissue Engineering, University of Manchester, Oxford Road, M13 9PL UK.
| | | | | |
Collapse
|