1
|
Hori A, Takahashi A, Miharu Y, Yamaguchi S, Sugita M, Mukai T, Nagamura F, Nagamura-Inoue T. Superior migration ability of umbilical cord-derived mesenchymal stromal cells (MSCs) toward activated lymphocytes in comparison with those of bone marrow and adipose-derived MSCs. Front Cell Dev Biol 2024; 12:1329218. [PMID: 38529405 PMCID: PMC10961348 DOI: 10.3389/fcell.2024.1329218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction: Mesenchymal stromal cells (MSCs) are activated upon inflammation and/or tissue damage and migrate to suppress inflammation and repair tissues. Migration is the first important step for MSCs to become functional; however, the migration potency of umbilical cord-derived MSCs (UC-MSCs) remains poorly understood. Thus, we aimed to assess the migration potency of UC-MSCs in comparison with those of bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) and investigate the influence of chemotactic factors on the migration of these cells. Methods: We compared the migration potencies of UC-, BM-, and AD-MSCs toward allogeneic stimulated mononuclear cells (MNCs) in mixed lymphocyte reaction (MLR). The number of MSCs in the upper chamber that migrated toward the MLR in the lower chamber was counted using transwell migration assay. Results and discussion: UC-MSCs showed significantly faster and higher proliferation potencies and higher migration potency toward unstimulated MNCs and MLR than BM- and AD-MSCs, although the migration potencies of the three types of MSCs were comparable when cultured in the presence of fetal bovine serum. The amounts of CCL2, CCL7, and CXCL2 in the supernatants were significantly higher in UC-MSCs co-cultured with MLR than in MLR alone and in BM- and AD-MSCs co-cultured with MLR, although they did not induce the autologous migration of UC-MSCs. The amount of CCL8 was higher in BM- and AD-MSCs than in UC-MSCs, and the amount of IP-10 was higher in AD-MSCs co-cultured with MLR than in UC- and BM-MSCs. The migration of UC-MSCs toward the MLR was partially attenuated by platelet-derived growth factor, insulin-like growth factor 1, and matrix metalloproteinase inhibitors in a dose-dependent manner. Conclusion: UC-MSCs showed faster proliferation and higher migration potency toward activated or non-activated lymphocytes than BM- and AD-MSCs. The functional chemotactic factors may vary among MSCs derived from different tissue sources, although the roles of specific chemokines in the different sources of MSCs remain to be resolved.
Collapse
Affiliation(s)
- Akiko Hori
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Somatic Stem Cell Research, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Somatic Stem Cell Research, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuta Miharu
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Somatic Stem Cell Research, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Masatoshi Sugita
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | - Takeo Mukai
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Fumitaka Nagamura
- Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Somatic Stem Cell Research, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Sun X, Lin Z, Xu N, Chen Y, Bian S, Zheng W. Identifying Dental Pulp Stem Cell as a Novel Therapeutic trategy for Digestive Diseases. Curr Stem Cell Res Ther 2024; 19:1293-1302. [PMID: 38018204 DOI: 10.2174/011574888x275737231120045815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 11/30/2023]
Abstract
Mesenchymal stem cells (MSCs) have been identified as potential therapeutics for various diseases. In contrast to other sources of MSCs, dental stem cells (DSCs) have received increased attention due to their high activity and easy accessibility. Among them, dental pulp stem cells (DPSCs) exhibit superior self-renewal, multipotency, immunomodulatory, and regenerative capacities. Following their inspiring performance in animal models and clinical trials, DPSCs show pharmacological potential in regenerative medicine. In this review, we have generalized the sources, heterogeneity, and biological characteristics of DPSCs, as well as compared them with other types of dental stem cells. In addition, we summarized the application of DPSCs in digestive diseases (such as liver, esophageal, and intestinal diseases), highlighting their regenerative and pharmacological potential based on the existing preclinical and clinical evidence. Specifically, DPSCs can be home to injured or inflamed tissues and exert repair and regeneration functions by facilitating immune regulation, anti-inflammation, and directional differentiation. Although DPSCs have a rosy prospect, future studies should handle the underlying drawbacks and pave the way for the identification of DPSCs as novel regenerative medicine.
Collapse
Affiliation(s)
- Xieyin Sun
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Zhaoyi Lin
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Nuo Xu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Yinqi Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Saiyan Bian
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
- Research Institute of Stem Cells, Center of Clinical Trials, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| |
Collapse
|
3
|
Ohnishi T, Homan K, Fukushima A, Ukeba D, Iwasaki N, Sudo H. A Review: Methodologies to Promote the Differentiation of Mesenchymal Stem Cells for the Regeneration of Intervertebral Disc Cells Following Intervertebral Disc Degeneration. Cells 2023; 12:2161. [PMID: 37681893 PMCID: PMC10486900 DOI: 10.3390/cells12172161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD), a highly prevalent pathological condition worldwide, is widely associated with back pain. Treatments available compensate for the impaired function of the degenerated IVD but typically have incomplete resolutions because of their adverse complications. Therefore, fundamental regenerative treatments need exploration. Mesenchymal stem cell (MSC) therapy has been recognized as a mainstream research objective by the World Health Organization and was consequently studied by various research groups. Implanted MSCs exert anti-inflammatory, anti-apoptotic, and anti-pyroptotic effects and promote extracellular component production, as well as differentiation into IVD cells themselves. Hence, the ultimate goal of MSC therapy is to recover IVD cells and consequently regenerate the extracellular matrix of degenerated IVDs. Notably, in addition to MSC implantation, healthy nucleus pulposus (NP) cells (NPCs) have been implanted to regenerate NP, which is currently undergoing clinical trials. NPC-derived exosomes have been investigated for their ability to differentiate MSCs from NPC-like phenotypes. A stable and economical source of IVD cells may include allogeneic MSCs from the cell bank for differentiation into IVD cells. Therefore, multiple alternative therapeutic options should be considered if a refined protocol for the differentiation of MSCs into IVD cells is established. In this study, we comprehensively reviewed the molecules, scaffolds, and environmental factors that facilitate the differentiation of MSCs into IVD cells for regenerative therapies for IDD.
Collapse
Affiliation(s)
- Takashi Ohnishi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Kentaro Homan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Akira Fukushima
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Daisuke Ukeba
- Department of Orthopedic Surgery, Hokkaido University Hospital, Sapporo 060-8648, Japan;
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
4
|
Mai Z, Chen H, Ye Y, Hu Z, Sun W, Cui L, Zhao X. Translational and Clinical Applications of Dental Stem Cell-Derived Exosomes. Front Genet 2021; 12:750990. [PMID: 34764982 PMCID: PMC8576041 DOI: 10.3389/fgene.2021.750990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising seed cells in tissue repair and regeneration due to their featured properties of self-renewal and multipotency. However, a growing body of evidence has demonstrated that MSCs exert biological functions mainly through secreting exosomes. Exosomes, which contain RNA, proteins, lipids, and metabolites, are new players in regulating many fundamental processes and play important roles in regenerative medicine. Exosomes not only mimic the effects of their parent cells but also possess many advantages such as high drug loading capacity, low immunogenicity, excellent biocompatibility, and low side effects. Currently, a total of 6 different dental stem cells (DSCs) including dental pulp stem cells (DPSCs), stem cells from exfoliated deciduous teeth (SHEDs), periodontal ligament stem cells (PDLSCs), dental follicle progenitor cells (DFPCs), stem cells from apical papilla (SCAPs) and gingival mesenchymal stem cells (GMSCs) have been isolated and identified. DSC-derived exosomes (DSC-Exos) are actively involved in intercellular communication, anti-inflammation, osteogenesis, angiogenesis, immunomodulation, nurturing neurons, and promoting tumor cell apoptosis. In this review, we will critically review the emerging role and clinical application potential of DSC-Exos.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Huan Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yu Ye
- Institute of Stomatology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Oral Diseases of Jiangsu Province, Stomatological Institute, Nanjing Medical University, Nanjing, China
| | - Ziyu Hu
- Department of Pediatrics, Nanjing Jinling Stomatology Hospital, Nanjing, China
| | - Wenjuan Sun
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Cui
- Stomatological Hospital, Southern Medical University, Guangzhou, China.,UCLA School of Dentistry, Los Angeles, CA, United States
| | - Xinyuan Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Panwar U, Mishra K, Patel P, Bharadva S, Vaniawala S, Shah A, Vundinti BR, Kothari SL, Ghosh K. Assessment of Long-Term in vitro Multiplied Human Wharton's Jelly-Derived Mesenchymal Stem Cells prior to Their Use in Clinical Administration. Cells Tissues Organs 2021; 210:239-249. [PMID: 34521091 DOI: 10.1159/000517423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/13/2021] [Indexed: 11/19/2022] Open
Abstract
The quantity of mesenchymal stem/stromal cells (MSCs) required for a particular therapy demands their subsequent expansion through ex vivo culture. During in vitro multiplication, they undergo replicative senescence which may alter their genetic stability. Therefore, this study was aimed to analyze cellular, molecular, and chromosomal alterations in Wharton's jelly-derived MSCs (WJ-MSCs) during their in vitro sequential passages, where WJ-MSCs were sequentially passaged up to P14 and cells were evaluated at an interval of P2, P6, P10, and P14. They were examined for their morphology, tumorigenicity, surface markers, stemness markers, DNA damage, chromosomal aberration, and telomere length. We have processed five full-term delivered human umbilical cord samples to obtain WJ-MSCs. Morphological appearance observed at initial stages was small fine spindle-shaped WJ-MSCs which were transformed to flat, long, and broader cells in later passages. The cell proliferation rate was gradually decreased after the 10th passage. WJ-MSCs have expressed stemness markers OCT-4 and NANOG, while they showed high expression of positive surface markers CD90 and CD105 and lower expression of CD34 and CD45. They were non-tumorigenic with slow cellular aging during subsequent passages. There was no chromosomal abnormality up to the 14th passage, while increase in comet score and decrease in telomere length were observed in later passages. Hence, our study suggests that early and middle passaged (less than P10) WJ-MSCs are good candidates for clinical administration for treatment.
Collapse
Affiliation(s)
- Urvi Panwar
- Surat Raktadan Kendra and Research Centre, Surat, India, .,Amity University Rajasthan, Jaipur, India,
| | | | - Parizad Patel
- Surat Raktadan Kendra and Research Centre, Surat, India
| | | | | | - Anjali Shah
- ICMR - National Institute of Immunohaematology, Mumbai, India
| | | | | | | |
Collapse
|
6
|
Mukai T, Sei K, Nagamura-Inoue T. Mesenchymal Stromal Cells Perspective: New Potential Therapeutic for the Treatment of Neurological Diseases. Pharmaceutics 2021; 13:pharmaceutics13081159. [PMID: 34452120 PMCID: PMC8401282 DOI: 10.3390/pharmaceutics13081159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/13/2022] Open
Abstract
Several studies have shown that mesenchymal stromal/stem cells (MSCs) exert their neuroprotective and neurorestorative efficacy via the secretion of neurotrophic factors. Based on these studies, many clinical trials using MSCs for the treatment of neurological disorders have been conducted, and results regarding their feasibility and efficacy have been reported. The present review aims to highlight the characteristics and basic research regarding the role of MSCs in neurological disease and to discuss the recent progress in clinical trials using MSCs to treat various neurological disorders.
Collapse
Affiliation(s)
- Takeo Mukai
- Department of Pediatrics, The University of Tokyo Hospital, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; (K.S.); (T.N.-I.)
- Correspondence: ; Tel.: +81-3-3815-5411; Fax: 81-3-5449-5452
| | - Kenshi Sei
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; (K.S.); (T.N.-I.)
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; (K.S.); (T.N.-I.)
| |
Collapse
|
7
|
Razavi M, Rezaee M, Telichko A, Inan H, Dahl J, Demirci U, Thakor AS. The Paracrine Function of Mesenchymal Stem Cells in Response to Pulsed Focused Ultrasound. Cell Transplant 2021; 29:963689720965478. [PMID: 33028105 PMCID: PMC7784560 DOI: 10.1177/0963689720965478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We studied the paracrine function of mesenchymal stem cells (MSCs) derived from various sources in response to pulsed focused ultrasound (pFUS). Human adipose tissue (AD), bone marrow (BM), and umbilical cord (UC) derived MSCs were exposed to pFUS at two intensities: 0.45 W/cm2 ISATA (310 kPa PNP) and 1.3 W/cm2 ISATA (540 kPa PNP). Following pFUS, the viability and proliferation of MSCs were assessed using a hemocytometer and confocal microscopy, and their secreted cytokine profile determined using a multiplex ELISA. Our findings showed that pFUS can stimulate the production of immunomodulatory, anti-inflammatory, and angiogenic cytokines from MSCs which was dependent on both the source of MSC being studied and the acoustic intensity employed. These important findings set the foundation for additional mechanistic and validation studies using this novel noninvasive and clinically translatable technology for modulating MSC biology.
Collapse
Affiliation(s)
- Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA.,BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, 6243University of Central Florida, Orlando, FL, USA.,Department of Materials Science and Engineering, 6243University of Central Florida, Orlando, FL, USA
| | - Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Arsenii Telichko
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Hakan Inan
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Jeremy Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Utkan Demirci
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, 6429Stanford University, Palo Alto, CA, USA
| |
Collapse
|
8
|
Umbilical cord-derived mesenchymal stromal cells immunomodulate and restore actin dynamics and phagocytosis of LPS-activated microglia via PI3K/Akt/Rho GTPase pathway. Cell Death Discov 2021; 7:46. [PMID: 33723246 PMCID: PMC7961004 DOI: 10.1038/s41420-021-00436-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 02/13/2021] [Indexed: 12/27/2022] Open
Abstract
Microglia are the immune cells in the central nervous system surveying environment and reacting to various injuries. Activated microglia may cause impaired synaptic plasticity, therefore modulating and restoring them to neutral phenotype is crucial to counteract a pro-inflammatory, neurotoxic state. In this study, we focused on elucidating whether human umbilical cord (UC) -derived mesenchymal stromal cells (MSCs) can exert immunomodulatory effect and change the phenotype of activated microglia. Primary culture of microglia was activated by lipopolysaccharide (LPS) and was co-cultured with three lots of MSCs. We investigated immunomodulation, actin dynamics and phagocytic capacity of activated microglia, and examined change of Rho GTPase in microglia as the mechanism. MSCs suppressed the expression of IL-1β and pNFκB in LPS-activated microglia, and conversely elevated the expression of IL-1β in resting-surveying microglia with lot-to-lot variation. Morphological and phagocytotic analyses revealed that LPS stimulation significantly increased active Rho GTPase, Rac1, and Cdc42 levels in the microglia, and their morphology changed to amoeboid in which F-actin spread with ruffle formation. The F-actin spreading persisted after removal of LPS stimulation and reduced phagocytosis. On the other hand, MSC co-culture induced bimodal increase in active Rac1 and Cdc42 levels in LPS-activated microglia. Moreover, extended ruffles of F-actin shrinked and concentrated to form an actin ring, thereby restoring phagocytosis. We confirmed inhibition of the PI3K/Akt pathway attenuated F-actin dynamics and phagocytosis restored by MSCs. Overall, we demonstrated that MSCs immunomodulated microglia with lot-to-lot variation, and changed the phenotype of LPS-activated microglia restoring actin dynamics and phagocytosis by increase of active Rho GTPase.
Collapse
|
9
|
Recent Updates of Diagnosis, Pathophysiology, and Treatment on Osteoarthritis of the Knee. Int J Mol Sci 2021; 22:ijms22052619. [PMID: 33807695 PMCID: PMC7961389 DOI: 10.3390/ijms22052619] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative and chronic joint disease characterized by clinical symptoms and distortion of joint tissues. It primarily damages joint cartilage, causing pain, swelling, and stiffness around the joint. It is the major cause of disability and pain. The prevalence of OA is expected to increase gradually with the aging population and increasing prevalence of obesity. Many potential therapeutic advances have been made in recent years due to the improved understanding of the underlying mechanisms, diagnosis, and management of OA. Embryonic stem cells and induced pluripotent stem cells differentiate into chondrocytes or mesenchymal stem cells (MSCs) and can be used as a source of injectable treatments in the OA joint cavity. MSCs are known to be the most studied cell therapy products in cell-based OA therapy owing to their ability to differentiate into chondrocytes and their immunomodulatory properties. They have the potential to improve cartilage recovery and ultimately restore healthy joints. However, despite currently available therapies and advances in research, unfulfilled medical needs persist for OA treatment. In this review, we focused on the contents of non-cellular and cellular therapies for OA, and briefly summarized the results of clinical trials for cell-based OA therapy to lay a solid application basis for clinical research.
Collapse
|
10
|
Monaco G, El Haj AJ, Alini M, Stoddart MJ. Ex Vivo Systems to Study Chondrogenic Differentiation and Cartilage Integration. J Funct Morphol Kinesiol 2021; 6:E6. [PMID: 33466400 PMCID: PMC7838775 DOI: 10.3390/jfmk6010006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Articular cartilage injury and repair is an issue of growing importance. Although common, defects of articular cartilage present a unique clinical challenge due to its poor self-healing capacity, which is largely due to its avascular nature. There is a critical need to better study and understand cellular healing mechanisms to achieve more effective therapies for cartilage regeneration. This article aims to describe the key features of cartilage which is being modelled using tissue engineered cartilage constructs and ex vivo systems. These models have been used to investigate chondrogenic differentiation and to study the mechanisms of cartilage integration into the surrounding tissue. The review highlights the key regeneration principles of articular cartilage repair in healthy and diseased joints. Using co-culture models and novel bioreactor designs, the basis of regeneration is aligned with recent efforts for optimal therapeutic interventions.
Collapse
Affiliation(s)
- Graziana Monaco
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
| | - Alicia J. El Haj
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
- Healthcare Technology Institute, Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
| |
Collapse
|
11
|
Al-Dhamin Z, Liu LD, Li DD, Zhang SY, Dong SM, Nan YM. Therapeutic efficiency of bone marrow-derived mesenchymal stem cells for liver fibrosis: A systematic review of in vivo studies. World J Gastroenterol 2020; 26:7444-7469. [PMID: 33384547 PMCID: PMC7754546 DOI: 10.3748/wjg.v26.i47.7444] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/31/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Although multiple drugs are accessible for recovering liver function in patients, none are considered efficient. Liver transplantation is the mainstay therapy for end-stage liver fibrosis. However, the worldwide shortage of healthy liver donors, organ rejection, complex surgery, and high costs are prompting researchers to develop novel approaches to deal with the overwhelming liver fibrosis cases. Mesenchymal stem cell (MSC) therapy is an emerging alternative method for treating patients with liver fibrosis. However, many aspects of this therapy remain unclear, such as the efficiency compared to conventional treatment, the ideal MSC sources, and the most effective way to use it. Because bone marrow (BM) is the largest source for MSCs, this paper used a systematic review approach to study the therapeutic efficiency of MSCs against liver fibrosis and related factors. We systematically searched multiple published articles to identify studies involving liver fibrosis and BM-MSC-based therapy. Analyzing the selected studies showed that compared with conventional treatment BM-MSC therapy may be more efficient for liver fibrosis in some cases. In contrast, the cotreatment presented a more efficient way. Nevertheless, BM-MSCs are lacking as a therapy for liver fibrosis; thus, this paper also reviews factors that affect BM-MSC efficiency, such as the implementation routes and strategies employed to enhance the potential in alleviating liver fibrosis. Ultimately, our review summarizes the recent advances in the BM-MSC therapy for liver fibrosis. It is grounded in recent developments underlying the efficiency of BM-MSCs as therapy, focusing on the preclinical in vivo experiments, and comparing to other treatments or sources and the strategies used to enhance its potential while mentioning the research gaps.
Collapse
Affiliation(s)
- Zaid Al-Dhamin
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| | - Ling-Di Liu
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| | - Dong-Dong Li
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| | - Si-Yu Zhang
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| | - Shi-Ming Dong
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| | - Yue-Min Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University & Hebei Key Laboratory of Mechanism of Liver Fibrosis in Chronic Liver Disease, Shijiazhuang 050051, Hebei Province, China
| |
Collapse
|
12
|
Stefańska K, Mehr K, Wieczorkiewicz M, Kulus M, Angelova Volponi A, Shibli JA, Mozdziak P, Skowroński MT, Antosik P, Jaśkowski JM, Piotrowska-Kempisty H, Kempisty B, Dyszkiewicz-Konwińska M. Stemness Potency of Human Gingival Cells-Application in Anticancer Therapies and Clinical Trials. Cells 2020; 9:cells9081916. [PMID: 32824702 PMCID: PMC7464983 DOI: 10.3390/cells9081916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/01/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Gingivae, as the part of periodontium, are involved in tooth support and possess the ability to heal rapidly, without scar formation. Recently, dental tissues have been identified as a potential source of mesenchymal stem cells (MSCs) and several populations of MSCs were isolated from the orofacial region, including gingival mesenchymal stem cells (GMSCs). GMSCs exhibit robust immunomodulatory and differentiation potential and are easily obtainable, which make them promising candidates for cellular therapies. Apart from being tested for application in immunologic- and inflammatory-related disorders and various tissue regeneration, GMSCs promise to be a valuable tool in cancer treatment, especially in tongue squamous cell carcinoma (TSCC) with the use of targeted therapy, since GMSCs are able to selectively migrate towards the cancerous cells both in vitro and in vivo. In addition to their ability to uptake and release anti-neoplastic drugs, GMSCs may be transduced with apoptosis-inducing factors and used for cancer growth inhibition. Moreover, GMSCs, as most mammalian cells, secrete exosomes, which are a subset of extracellular vesicles with a diameter of 40–160 nm, containing DNA, RNA, lipids, metabolites, and proteins. Such GMSCs-derived exosomes may be useful therapeutic tool in cell-free therapy, as well as their culture medium. GMSCs exhibit molecular and stem-cell properties that make them well suited in preclinical and clinical studies.
Collapse
Affiliation(s)
- Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland;
| | - Katarzyna Mehr
- Department of Gerostomatology and Pathology of Oral Cavity, Poznan University of Medical Sciences, 70 Bukowska St., 60-812 Poznan, Poland;
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland; (M.W.); (M.T.S.)
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland; (M.K.); (P.A.)
| | - Ana Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, Dental Institute, King’s College London, Strand, London WC2R 2LS, UK;
| | - Jamil A. Shibli
- Department of Periodontology and Oral Implantology, Dental Research Division, Guarulhos University, Guarulhos, R. Eng. Prestes Maia, 88-Centro, São Paulo 07023-070, Brazil;
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Campus Box 7608, Raleigh, NC 27695-7608, USA;
| | - Mariusz T. Skowroński
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland; (M.W.); (M.T.S.)
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland; (M.K.); (P.A.)
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 7 Gagarina St., 87-100 Torun, Poland; (M.K.); (P.A.)
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland;
- Correspondence: ; Tel./Fax: +48-61-8546565
| | - Marta Dyszkiewicz-Konwińska
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland;
- Department of Biomaterials and Experimental Dentistry, Poznan University of Medical Sciences, 70 Bukowska St., 60-812 Poznan, Poland
| |
Collapse
|
13
|
Human Wharton's Jelly-Cellular Specificity, Stemness Potency, Animal Models, and Current Application in Human Clinical Trials. J Clin Med 2020; 9:jcm9041102. [PMID: 32290584 PMCID: PMC7230974 DOI: 10.3390/jcm9041102] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stem cell therapies offer a great promise for regenerative and reconstructive medicine, due to their self-renewal and differentiation capacity. Although embryonic stem cells are pluripotent, their utilization involves embryo destruction and is ethically controversial. Therefore, adult tissues that have emerged as an alternative source of stem cells and perinatal tissues, such as the umbilical cord, appear to be particularly attractive. Wharton's jelly, a gelatinous connective tissue contained in the umbilical cord, is abundant in mesenchymal stem cells (MSCs) that express CD105, CD73, CD90, Oct-4, Sox-2, and Nanog among others, and have the ability to differentiate into osteogenic, adipogenic, chondrogenic, and other lineages. Moreover, Wharton's jelly-derived MSCs (WJ-MSCs) do not express MHC-II and exhibit immunomodulatory properties, which makes them a good alternative for allogeneic and xenogeneic transplantations in cellular therapies. Therefore, umbilical cord, especially Wharton's jelly, is a promising source of mesenchymal stem cells.
Collapse
|
14
|
Sepúlveda RV, Eleotério In Memorian RB, Valente FL, Araújo FR, Sabino ADP, Evangelista FCG, Reis ECC, Borges APB. Canine umbilical cord perivascular tissue: A source of stem cells for therapy and research. Res Vet Sci 2020; 129:193-202. [PMID: 32087438 DOI: 10.1016/j.rvsc.2020.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/17/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
There are numerous sources of multipotent mesenchymal stromal cells (MSC) with therapeutic potential, and bone marrow is the main one. However, pain, lack of donors and comorbidities associated with harvesting stimulate the search for new sources of MSCs. The aim of this work is to obtain cells from umbilical cord (UC) perivascular tissue of dogs and characterize them as MSCs. For this, the UC was obtained from therapeutic cesarean sections and submitted to enzymatic digestion. The obtained cells were subjected to growth and proliferation tests, as well as the analysis of surface markers, differentiation test in three mesenchymal lineages and analysis of differentiation markers expression. From all the UC used in this study an adherent with fibroblastoid shape cell was obtained, with an initial number of 4.8 × 105 of cells. The growth curves showed a lag phase from 0 to 24 h, followed by a phase of growth of 24 to 168 h, and then phase of cell decay. The doubling time was kept around 15 h until the sixth passage, from which there were signs of cellular senescence. The differentiation assays demonstrated the ability of cells to differentiate into osteoblasts, adipocytes and chondrocytes when subjected to the induction mediums. The study of surface markers was positive for adhesion markers and negative for hematopoietic markers. Thus, cells obtained from canine UC perivascular tissue by enzymatic digestion are multipotent MSC and the protocol developed ensures the perivascular origin of these cells.
Collapse
Affiliation(s)
| | | | | | - Fabiana Rocha Araújo
- Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Adriano de Paula Sabino
- Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | | | | | | |
Collapse
|
15
|
Chirumbolo S. Oxidative Stress, Nutrition and Cancer: Friends or Foes? World J Mens Health 2020; 39:19-30. [PMID: 32202081 PMCID: PMC7752511 DOI: 10.5534/wjmh.190167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
The relationship between cancer and nutrition, as well as nutrition and oxidative stress, shares puzzling aspects that current research is investigating as the possible components of an intriguing regulating mechanism involving the complex interplay between adipose tissue and other compartments. Along the very recent biological evolution, humans underwent a rapid change in their lifestyles and henceforth the role of the adipocytes earned a much more complex task in the fine tuning of the tissue microenvironment. A lipidic signaling language probably evolved in association with the signaling role of reactive oxygen species, which gained a fundamental part in the regulation of cell stem and plasticity. The possible relationship with cancer onset might have some causative mechanism in the impairment of this complex task, usually deregulated by drastic changes in one's own lifestyle and dietary habit. This review tries to address this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
16
|
Zhou Y, Yamamoto Y, Xiao Z, Ochiya T. The Immunomodulatory Functions of Mesenchymal Stromal/Stem Cells Mediated via Paracrine Activity. J Clin Med 2019; 8:jcm8071025. [PMID: 31336889 PMCID: PMC6678920 DOI: 10.3390/jcm8071025] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) exist in almost all tissues, possessing the potential to differentiate into specialized cell types and exert immunomodulatory functions. Thus, they have attracted much attention as a promising therapeutic candidate. Recent studies have demonstrated that paracrine signaling is mainly responsible for the involvement of MSCs in the modulation of immune responses and the progression of diseases. Through release of secretome consisting of a diverse range of cytokines, chemokines, and extracellular vesicles (EVs), MSCs convey regulatory messages to recipient immune cells in the microenvironment. In this review, we focus on the recent advances in how MSCs contribute to immunomodulation through the secretion of paracrine factors. The further improved understanding of the molecular mechanism underlying the interactions between MSCs and immune cells highlights the paracrine biology of MSCs in the modulation of the immune microenvironment and promotes the clinical application of MSCs in regenerative medicine and immune diseases.
Collapse
Affiliation(s)
- Yueyuan Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Division of Cellular Signaling, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Takahiro Ochiya
- Division of Cellular Signaling, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan.
| |
Collapse
|
17
|
Mukai T, Tojo A, Nagamura-Inoue T. Mesenchymal stromal cells as a potential therapeutic for neurological disorders. Regen Ther 2018; 9:32-37. [PMID: 30525073 PMCID: PMC6222283 DOI: 10.1016/j.reth.2018.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Several studies have reported that mesenchymal stromal/stem cells (MSCs) restore neurological damage through their secretion of paracrine factors or their differentiation to neuronal cells. Based on these studies, many clinical trials have been conducted using MSCs for neurological disorders, and their safety and efficacy have been reported. In this review, we provide a brief introduction to MSCs, especially umbilical cord derived-MSCs (UC-MSCs), in terms of characteristics, isolation, and cryopreservation, and discuss the recent progress in regenerative therapies using MSCs for various neurological disorders.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.,Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.,Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| |
Collapse
|
18
|
Mukai T, Tojo A, Nagamura-Inoue T. Umbilical Cord-Derived Mesenchymal Stromal Cells Contribute to Neuroprotection in Neonatal Cortical Neurons Damaged by Oxygen-Glucose Deprivation. Front Neurol 2018; 9:466. [PMID: 29963009 PMCID: PMC6013549 DOI: 10.3389/fneur.2018.00466] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
Several studies have reported that human umbilical cord-derived mesenchymal stromal cells (UC-MSCs) restore neurological damage in vivo through their secretion of paracrine factors. We previously found that UC-MSCs attenuate brain injury by secreting neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and hepatocyte growth factor (HGF). However, how these factors contribute to neuroprotection remains unknown. In this study, we aimed to investigate to what extent UC-MSC-derived HGF and BDNF contribute to neuroprotection using a Transwell co-culture system of neonatal cortical neurons damaged by oxygen-glucose deprivation. The influence of HGF and BDNF were determined by investigating neurons in both the presence and absence of UC-MSCs as these cells consistently secrete both factors and can be blocked by neutralizing antibodies. In the co-culture, UC-MSCs significantly improved neuronal injury, as indicated by an increase in immature neuron number, neurite outgrowth, and cell proliferation. Co-culture of damaged neurons with UC-MSCs also exhibited a reduction in the number of neurons displaying signs of apoptosis/necrosis. The neuroprotective actions of UC-MSCs were partially reverted by neutralizing antibodies. Together, our findings reveal that UC-MSC-secreted HGF and BDNF have neuroprotective effects on damaged neurons. Further studies should address the existence of other potential neurotrophic paracrine factors.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Liu C, Li T, Yang Z, Liu D, Li Y, Zhou Z, Zhang Q. Kartogenin Enhanced Chondrogenesis in Cocultures of Chondrocytes and Bone Mesenchymal Stem Cells. Tissue Eng Part A 2018; 24:990-1000. [DOI: 10.1089/ten.tea.2017.0162] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Chun Liu
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| | - Tao Li
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| | - Zhijian Yang
- Agricultural Product Quality Institute, Fujian Agriculture and Forestry University, Fuzhou, P.R. China
| | - Deshuai Liu
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| | - Yun Li
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| | - Zhiyou Zhou
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| | - Qiqing Zhang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou, P.R. China
| |
Collapse
|
20
|
Mussano F, Genova T, Petrillo S, Roato I, Ferracini R, Munaron L. Osteogenic Differentiation Modulates the Cytokine, Chemokine, and Growth Factor Profile of ASCs and SHED. Int J Mol Sci 2018; 19:ijms19051454. [PMID: 29757956 PMCID: PMC5983594 DOI: 10.3390/ijms19051454] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023] Open
Abstract
Great efforts have been made to improve bone regeneration techniques owing to a growing variety of sources of stem cells suitable for autologous transplants. Specifically, adipose-derived stem cells (ASCs) and stems cells from human exfoliated deciduous teeth (SHED) hold great potential for bone tissue engineering and cell therapy. After a preliminary characterization of the main biomolecules ASCs and SHED released in their conditioned media, cells were kept both in normal and osteo-inducing conditions. Conventional assays were performed to prove their osteogenic potential such as quantitative real-time polymerase chain reaction (qRT-PCR) (for RUNX-2, collagen type I, osteopontin and osteonectin), alkaline phosphatase activity, osteocalcin production, and von Kossa staining. Conditioned media were tested again after the osteogenic induction and compared to maintaining condition both at base line and after 14 days of culture. The osteogenic condition inhibited the release of all the biomolecules, with the exception, concerning SHED, of growth-regulated alpha protein precursor (GROα), and, to a lesser extent, interleukin (IL)-8. In conclusion, our data support that undifferentiated ASCs and SHED may be preferable to committed ones for general cell therapy approaches, due to their higher paracrine activity. Osteoinduction significantly affects the cytokine, chemokine, and growth factor profile in a differential way, as SHED kept a more pronounced pro-angiogenic signature than ASCs.
Collapse
Affiliation(s)
- Federico Mussano
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
| | - Tullio Genova
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, UNITO, Via Nizza 52, 10126 Turin, Italy.
| | - Ilaria Roato
- Center for Research and Medical Studies, A.O.U. Città della Salute e della Scienza, 10126 Turin, Italy.
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genoa, Italy.
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
21
|
Human Urine-Derived Stem Cells: Potential for Cell-Based Therapy of Cartilage Defects. Stem Cells Int 2018; 2018:4686259. [PMID: 29765413 PMCID: PMC5932456 DOI: 10.1155/2018/4686259] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/25/2018] [Accepted: 03/19/2018] [Indexed: 02/05/2023] Open
Abstract
Stem cell therapy is considered an optimistic approach to replace current treatments for cartilage defects. Recently, human urine-derived stem cells (hUSCs), which are isolated from the urine, are studied as a promising candidate for many tissue engineering therapies due to their multipotency and sufficient proliferation activities. However, it has not yet been reported whether hUSCs can be employed in cartilage defects. In this study, we revealed that induced hUSCs expressed chondrogenic-related proteins, including aggrecan and collagen II, and their gene expression levels were upregulated in vitro. Moreover, we combined hUSCs with hyaluronic acid (HA) and injected hUSCs-HA into a rabbit knee joint with cartilage defect. Twelve weeks after the injection, the histologic analyses (HE, toluidine blue, and Masson trichrome staining), immunohistochemistry (aggrecan and collagen II), and histologic grade of the sample indicated that hUSCs-HA could stimulate much more neocartilage formation compared with hUSCs alone, pure HA, and saline, which only induced the modest cartilage regeneration. In this study, we demonstrated that hUSCs could be a potential cell source for stem cell therapies to treat cartilage-related defects in the future.
Collapse
|
22
|
Elsaadany M, Winters K, Adams S, Stasuk A, Ayan H, Yildirim-Ayan E. Equiaxial Strain Modulates Adipose-derived Stem Cell Differentiation within 3D Biphasic Scaffolds towards Annulus Fibrosus. Sci Rep 2017; 7:12868. [PMID: 28993681 PMCID: PMC5634474 DOI: 10.1038/s41598-017-13240-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/19/2017] [Indexed: 12/14/2022] Open
Abstract
Recurrence of intervertebral disc (IVD) herniation is the most important factor leading to chronic low back pain and subsequent disability after discectomy. Efficacious annulus fibrosus (AF) repair strategy that delivers cells and biologics to IVD injury site is needed to limit the progression of disc degeneration and promote disc self-regeneration capacities after discectomy procedures. In this study, a biphasic mechanically-conditioned scaffold encapsulated with human adipose-derived stem cells (ASCs) is studied as a potential treatment strategy for AF defects. Equiaxial strains and frequencies were applied to ASCs-encapsulated scaffolds to identify the optimal loading modality to induce AF differentiation. Equiaxial loading resulted in 2–4 folds increase in secretion of extracellular matrix proteins and the reorganization of the matrix fibers and elongations of the cells along the load direction. Further, the equiaxial load induced region-specific differentiation of ASCs within the inner and outer regions of the biphasic scaffolds. Gene expression of AF markers was upregulated with 5–30 folds within the equiaxially loaded biphasic scaffolds compared to unstrained samples. The results suggest that there is a specific value of equiaxial strain favorable to differentiate ASCs towards AF lineage and that ASCs-embedded biphasic scaffold can potentially be utilized to repair the AF defects.
Collapse
Affiliation(s)
| | - Kayla Winters
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Sarah Adams
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Alexander Stasuk
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Halim Ayan
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
23
|
Intravenous injection of umbilical cord-derived mesenchymal stromal cells attenuates reactive gliosis and hypomyelination in a neonatal intraventricular hemorrhage model. Neuroscience 2017; 355:175-187. [PMID: 28504197 DOI: 10.1016/j.neuroscience.2017.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/21/2017] [Accepted: 05/03/2017] [Indexed: 12/26/2022]
Abstract
Intraventricular hemorrhage (IVH) is a frequent complication of preterm newborns, resulting in cerebral palsy and cognitive handicap as well as hypoxic ischemic encephalopathy and periventricular leukomalacia. In this study, we investigated the restorative effect on neonatal IVH by umbilical cord-derived mesenchymal stromal cells (UC-MSCs) cultured in serum-free medium (RM medium) for clinical application. UC-MSCs were cultured with αMEM medium supplemented with FBS or RM. A neonatal IVH mouse model at postnatal day 5 was generated by intraventricular injection of autologous blood, and mice were intravenously administered 1×105 UC-MSCs two days after IVH. Brain magnetic resonance imaging was performed at postnatal day 15, 22 and neurological behavioral measurements were performed at postnatal day 23, accompanied by histopathological analysis and cytokine bead assays in serum after IVH with or without UC-MSCs. Both UC-MSCs cultured with αMEM and RM met the criteria of MSCs and improved behavioral outcome of IVH mice. Moreover the RM group exhibited significant behavioral improvement compared to the control group. Histopathological analysis revealed UC-MSCs cultured with RM significantly attenuated periventricular reactive gliosis, hypomyelination, and periventricular cell death observed after IVH. Furthermore, human brain-derived neurotrophic factor and hepatocyte growth factor were elevated in the serum, cerebrospinal fluid and brain tissue of neonatal IVH model mice 24h after UC-MSCs administration. These results suggest UC-MSCs attenuate neonatal IVH by protecting gliosis and apoptosis of the injured brain, and intravenous injection of UC-MSCs cultured in RM may be feasible for neonatal IVH in clinic.
Collapse
|
24
|
Mukai T, Nagamura-Inoue T, Shimazu T, Mori Y, Takahashi A, Tsunoda H, Yamaguchi S, Tojo A. Neurosphere formation enhances the neurogenic differentiation potential and migratory ability of umbilical cord-mesenchymal stromal cells. Cytotherapy 2016; 18:229-41. [PMID: 26794714 DOI: 10.1016/j.jcyt.2015.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/02/2015] [Accepted: 10/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS The human umbilical cord (UC) is a rich source of mesenchymal stromal cells (MSCs), which have been reported to have multi-lineage potential. The objectives of this study were to investigate the characteristics and capacity of UC-MSC neurosphere formation and whether this event enhances the propensity of UC-MSCs to undergo neural differentiation. METHODS UC-MSCs were collected by the improved explant method. UC-MSCs and neurosphere-forming UC-MSCs (UC-MSC-neurospheres) were induced to undergo neurogenic differentiation, the latter of which were induced by suspension culturing in the presence of epidermal growth factor and basic fibroblast growth factor. The differentiation and migratory capacities of the individual cultures were then compared on the basis of the expression of neural markers, as measured by immunocytochemistry, immunoblotting and quantitative real-time polymerase chain reaction and transwell assays, respectively. RESULTS Both UC-MSCs and UC-MSC-neurospheres were capable of differentiating into neurogenic cells when cultured in neurogenic differentiation medium. However, pre-conditioned UC-MSC-neurospheres exhibited significantly higher expression of neural markers--including microtubule-associated protein (MAP2), MUSASHI1, glial fibrillary acidic protein (GFAP), and NESTIN--compared with those derived from UC-MSCs directly. Moreover, UC-MSC-neurospheres expressed significantly higher levels of the stemness markers NANOG, KLF4 and OCT4 than did UC-MSCs. Migration assays also revealed that both UC-MSCs and UC-MSC-neurospheres actively migrate toward glucose-depleted cells. CONCLUSIONS Neurogenic differentiation potential probably is greater in UC-MSC-neurospheres than in UC-MSCs. Thus, UC-MSC-neurospheres may serve as a better source of cells for neurogenic regenerative medicine.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | | | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Gruber HE, Riley FE, Hoelscher GL, Ingram JA, Bullock L, Hanley EN. Human annulus progenitor cells: Analyses of this viable endogenous cell population. J Orthop Res 2016; 34:1351-60. [PMID: 27249627 DOI: 10.1002/jor.23319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/27/2016] [Indexed: 02/04/2023]
Abstract
Back pain and intervertebral disc degeneration have growing socioeconomic/health care impacts. Increasing research efforts address use of stem and progenitor cell-based replacement therapies to repopulate and regenerate the disc. Data presented here on the innate human annulus progenitor cells: (i) assessed osteogenic, chondrogenic and adipogenic potentials of cultured human annulus cells; and (ii) defined progenitor-cell related gene expression patterns. Verification of the presence of progenitor cells within primary human disc tissue also used immunohistochemical identification of cell surface markers and microarray analyses. Differentiation analysis in cell cultures demonstrated a viable progenitor cell pool within Thompson grades III-IV discs. Osteogenesis was present in 8 out of 11 cultures (73%), chondrogenesis in 8 of 11 (73%), and adipogenesis in 6 of 6 (100%). Immunolocalization was positive for CD29, CD44, CD105, and CD14 (mean values 80.2%, 81.5%, 85.1%, and 88.6%, respectively); localization of CD45 and CD34 was negative in disc tissue. Compared to controls, surgical discs showed significantly downregulated genes with recognized progenitor cell functions: TCF7L2 (2.7 fold), BMI1 (3.8 fold), FGF receptor 2 (2 fold), PAFAH1B1 (2.3 fold), and GSTP1 (9 fold). Compared to healthier grade I/II discs, grade III/IV discs showed significantly upregulated XRCC5 (3.6 fold), TCF7L2 (6 fold), GSTP1 (3.7 fold), and BMI1 (3 fold). Additional significant cell marker analyses showed expression of platelet-derived growth factor receptor alpha, CD90, CD73, and STRO-1. Statement of Clinical Significance: Findings provide the first identification of progenitor cells in annulus specimens from older, more degenerate discs (in contrast to earlier studies of healthier discs or nondegenerative specimens from teenagers). Findings also increase knowledge on progenitor cells present in the disc and suggest their value in potential future utilization for regeneration and disc cell therapy. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1351-1360, 2016.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Frank E Riley
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Gretchen L Hoelscher
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Jane A Ingram
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Letitia Bullock
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Edward N Hanley
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| |
Collapse
|
26
|
Blanquer SB, Gebraad AW, Miettinen S, Poot AA, Grijpma DW, Haimi SP. Differentiation of adipose stem cells seeded towards annulus fibrosus cells on a designed poly(trimethylene carbonate) scaffold prepared by stereolithography. J Tissue Eng Regen Med 2016; 11:2752-2762. [DOI: 10.1002/term.2170] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 01/06/2016] [Accepted: 02/10/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Sébastien B.G. Blanquer
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
- Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation; Davos Switzerland
| | - Arjen W.H. Gebraad
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
- Department of Oral and Maxillofacial Sciences, Clinicum; University of Helsinki; Helsinki Finland
| | - Susanna Miettinen
- Institute of Biosciences and Medical Technology (BioMediTech); University of Tampere; Tampere Finland
| | - André A. Poot
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
- Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation; Davos Switzerland
| | - Dirk W. Grijpma
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
- Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation; Davos Switzerland
- University of Groningen, University Medical Centre Groningen, W.J. Kolff Institute; Department of Biomedical Engineering; Groningen the Netherlands
| | - Suvi P. Haimi
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Biomaterials Science and Technology; University of Twente; Enschede the Netherlands
- Department of Oral and Maxillofacial Sciences, Clinicum; University of Helsinki; Helsinki Finland
| |
Collapse
|
27
|
Manchikanti L, Boswell MV, Hirsch JA. Innovations in interventional pain management of chronic spinal pain. Expert Rev Neurother 2016; 16:1033-42. [DOI: 10.1080/14737175.2016.1194204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
28
|
Abstract
One of the most important issues facing cartilage tissue engineering is the inability to move technologies into the clinic. Despite the multitude of current research in the field, it is known that 90% of new drugs that advance past animal studies fail clinical trials. The objective of this review is to provide readers with an understanding of the scientific details of tissue engineered cartilage products that have demonstrated a certain level of efficacy in humans, so that newer technologies may be developed upon this foundation. Compared to existing treatments, such as microfracture or autologous chondrocyte implantation, a tissue engineered product can potentially provide more consistent clinical results in forming hyaline repair tissue and in filling the entirety of the defect. The various tissue engineering strategies (e.g., cell expansion, scaffold material, media formulations, biomimetic stimuli, etc.) used in forming these products, as collected from published literature, company websites, and relevant patents, are critically discussed. The authors note that many details about these products remain proprietary, not all information is made public, and that advancements to the products are continuously made. Nevertheless, by understanding the design and production processes of these emerging technologies, one can gain tremendous insight into how to best use them and also how to design the next generation of tissue engineered cartilage products.
Collapse
|
29
|
Huang BJ, Hu JC, Athanasiou KA. Cell-based tissue engineering strategies used in the clinical repair of articular cartilage. Biomaterials 2016; 98:1-22. [PMID: 27177218 DOI: 10.1016/j.biomaterials.2016.04.018] [Citation(s) in RCA: 270] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/15/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022]
Abstract
One of the most important issues facing cartilage tissue engineering is the inability to move technologies into the clinic. Despite the multitude of current research in the field, it is known that 90% of new drugs that advance past animal studies fail clinical trials. The objective of this review is to provide readers with an understanding of the scientific details of tissue engineered cartilage products that have demonstrated a certain level of efficacy in humans, so that newer technologies may be developed upon this foundation. Compared to existing treatments, such as microfracture or autologous chondrocyte implantation, a tissue engineered product can potentially provide more consistent clinical results in forming hyaline repair tissue and in filling the entirety of the defect. The various tissue engineering strategies (e.g., cell expansion, scaffold material, media formulations, biomimetic stimuli, etc.) used in forming these products, as collected from published literature, company websites, and relevant patents, are critically discussed. The authors note that many details about these products remain proprietary, not all information is made public, and that advancements to the products are continuously made. Nevertheless, by understanding the design and production processes of these emerging technologies, one can gain tremendous insight into how to best use them and also how to design the next generation of tissue engineered cartilage products.
Collapse
Affiliation(s)
- Brian J Huang
- Department of Biomedical Engineering, University of California Davis, USA.
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, USA.
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Davis, USA; Department of Orthopedic Surgery, University of California Davis, USA.
| |
Collapse
|
30
|
Gruber HE, Ode G, Hoelscher G, Ingram J, Bethea S, Bosse MJ. Osteogenic, stem cell and molecular characterisation of the human induced membrane from extremity bone defects. Bone Joint Res 2016; 5:106-15. [PMID: 27056768 PMCID: PMC5009235 DOI: 10.1302/2046-3758.54.2000483] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
Objectives The biomembrane (induced membrane) formed around polymethylmethacrylate (PMMA) spacers has value in clinical applications for bone defect reconstruction. Few studies have evaluated its cellular, molecular or stem cell features. Our objective was to characterise induced membrane morphology, molecular features and osteogenic stem cell characteristics. Methods Following Institutional Review Board approval, biomembrane specimens were obtained from 12 patient surgeries for management of segmental bony defects (mean patient age 40.7 years, standard deviation 14.4). Biomembranes from nine tibias and three femurs were processed for morphologic, molecular or stem cell analyses. Gene expression was determined using the Affymetrix GeneChip Operating Software (GCOS). Molecular analyses compared biomembrane gene expression patterns with a mineralising osteoblast culture, and gene expression in specimens with longer spacer duration (> 12 weeks) with specimens with shorter durations. Statistical analyses used the unpaired student t-test (two tailed; p < 0.05 was considered significant). Results Average PMMA spacer in vivo time was 11.9 weeks (six to 18). Trabecular bone was present in 33.3% of the biomembrane specimens; bone presence did not correlate with spacer duration. Biomembrane morphology showed high vascularity and collagen content and positive staining for the key bone forming regulators, bone morphogenetic protein 2 (BMP2) and runt-related transcription factor 2 (RUNX2). Positive differentiation of cultured biomembrane cells for osteogenesis was found in cells from patients with PMMA present for six to 17 weeks. Stem cell differentiation showed greater variability in pluripotency for osteogenic potential (70.0%) compared with chondrogenic or adipogenic potentials (100% and 90.0%, respectively). Significant upregulation of BMP2 and 6, numerous collagens, and bone gla protein was present in biomembrane compared with the cultured cell line. Biomembranes with longer resident PMMA spacer duration (vs those with shorter residence) showed significant upregulation of bone-related, stem cell, and vascular-related genes. Conclusion The biomembrane technique is gaining favour in the management of complicated bone defects. Novel data on biological mechanisms provide improved understanding of the biomembrane’s osteogenic potential and molecular properties. Cite this article: Dr H. E. Gruber. Osteogenic, stem cell and molecular characterisation of the human induced membrane from extremity bone defects. Bone Joint Res 2016;5:106–115. DOI: 10.1302/2046-3758.54.2000483.
Collapse
Affiliation(s)
- H E Gruber
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - G Ode
- Department of Orthopaedic Surgery, Carolinas Medical Center, Morehead Medical Plaza, 1025 Morehead Medical Drive, Suite 300, Charlotte, NC 28204, USA
| | - G Hoelscher
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232 USA
| | - J Ingram
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - S Bethea
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Research Center, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - M J Bosse
- Department of Orthopaedic Surgery, Morehead Medical Plaza, 1025 Morehead Medical Drive, Suite 300, Charlotte, NC 28204, USA
| |
Collapse
|
31
|
Shimazu T, Mori Y, Takahashi A, Tsunoda H, Tojo A, Nagamura-Inoue T. Serum- and xeno-free cryopreservation of human umbilical cord tissue as mesenchymal stromal cell source. Cytotherapy 2016; 17:593-600. [PMID: 25881518 DOI: 10.1016/j.jcyt.2015.03.604] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/02/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AIMS Human umbilical cord (UC) has become a notable source for mesenchymal stromal cells (MSCs) that can migrate to areas of inflammation and damaged tissue and can suppress excess immune reactions and to repair, respectively. Although UC is a solid tissue, there are several advantages, including repeatable uses from the same donor sample when needed and the possibility of future explorations for cells with unknown potential, if we could cryopreserve the UC as a living tissue material. However, because the cryoprotectants in the previous reports included animal- or allogeneic human-derived serum or no serum, the frozen-thawed UC-MSCs were inferior to fresh UC-MSCs in cell proliferation. The objective of this study was to find a suitable cryopreservation method of UC for clinical use. METHODS The UC was cut in cross-section and incised longitudinally, immersed in the cryoprotectant and frozen slowly. Later, it was thawed and minced rapidly, and the fragments of UC were cultured by improved explant method. RESULTS The highest yield of cells was obtained from frozen-thawed UC with serum- and xeno-free cryoprotectant, STEM-CELLBANKER, when compared with others. The cells derived from frozen-thawed UC stored in STEM-CELLBANKER expressed the phenotypes of MSCs, retained the immunosuppressive properties in allogeneic mixed lymphocyte reactions and the differentiation potentials (into adipocyte and chondrocytes) comparable to those derived from fresh UC. CONCLUSIONS UC can be cryopreserved in serum- and xeno-free cryoprotectant as a living tissue while keeping its growth and functions equivalent to fresh UC. Our method is simple and feasible for clinical use.
Collapse
Affiliation(s)
- Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | - Arinobu Tojo
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
32
|
Cellular supplementation technologies for painful spine disorders. PM R 2016; 7:S19-S25. [PMID: 25864656 DOI: 10.1016/j.pmrj.2015.01.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 11/23/2022]
Abstract
Low back pain affects more than 80% of adults. A proportion of these adults develops chronic low back pain (CLBP) and becomes disabled by their condition. CLBP is expensive to diagnose and treat and in terms of associated loss of productivity in the work place setting by affected individuals. Although challenging, the source of CLBP can be identified. Contemporary literature contains several studies that have established prevalence estimates for various structural sources of CLBP. In young adults, the intervertebral disk is a common source of CLBP, once it incurs annular injury that heals incompletely. Effective treatment for painful disks currently is an unmet clinical need. In older adults, the facet and sacroiliac joints are more commonly responsible for CLBP. Although certain minimally invasive techniques do exist for these painful joints, an effective restorative intervention has yet to be established. Annular injury precipitates a physiologic response that can lead to a catabolic state within the disk that impairs disk restoration. Cell loss is a feature of this process as well as the pathophysiology associated with painful facet and sacroiliac joints. Cellular supplementation is an attractive treatment strategy to initiate the repair of an injured lumbosacral structure. The introduction of exogenous cells may lead to increased extracelluar matrix production and reduced pain and disability in diskogenic CLBP. Compelling data in animal studies have been produced, stimulating Food and Drug Administration-regulated trials in humans. Numerous questions remain regarding cell viability and sufficient native nutrients to support these cells. Clinical research protocols have focused predominantly on diskogenic CLBP, and very few have addressed painful facet and/or sacroiliac joints.
Collapse
|
33
|
Mori Y, Ohshimo J, Shimazu T, He H, Takahashi A, Yamamoto Y, Tsunoda H, Tojo A, Nagamura-Inoue T. Improved Explant Method to Isolate Umbilical Cord-Derived Mesenchymal Stem Cells and Their Immunosuppressive Properties. Tissue Eng Part C Methods 2015; 21:367-72. [DOI: 10.1089/ten.tec.2014.0385] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun Ohshimo
- R & D Center, Tsubakimoto Chain Corporation, Saitama, Japan
| | - Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haiping He
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuki Yamamoto
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | - Arinobu Tojo
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Molecular Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Chen D, Hao H, Tong C, Liu J, Dong L, Ti D, Hou Q, Liu H, Han W, Fu X. Transdifferentiation of Umbilical Cord–Derived Mesenchymal Stem Cells Into Epidermal-Like Cells by the Mimicking Skin Microenvironment. INT J LOW EXTR WOUND 2015; 14:136-45. [PMID: 25700709 DOI: 10.1177/1534734615569913] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human umbilical cord–derived mesenchymal stem cells (UC-MSCs) are multipotent, primitive, and have been widely used for skin tissue engineering. Their transdifferentiation is determined by the local microenvironment. In this study, we investigated the potential epidermal differentiation of UC-MSCs and the formation of epidermis substitutes in a 3-dimensional (3D) microenvironment, which was fabricated by UC-MSCs embedded into collagen–chitosan scaffolds (CCSs) combined with an air–liquid interface (ALI) culture system. Using fluorescence microscope, we observed that UC-MSCs were spindle-shaped and evenly distributed in the scaffold. Methyl thiazolyl blue tetrazolium bromide assay and Live/Dead assay indicated that the CCSs have good biocompatibility with UC-MSCs. Immunohistochemistry and western blotting assay showed that UC-MSCs on the surface of the CCSs were positive for the epidermal markers cytokeratin 19 and involucrin at 14 days. In addition, hematoxylin–eosin staining indicated that multilayered epidermis substitutes were established. The constructed epidermis substitutes were applied to treat full-thickness wounds in rats and proved to promote wound healing. In conclusion, manipulating the 3D microenvironment is a novel method for inducing the epidermal differentiation of MSCs to engineer epidermal substitutes, which provides an alternative strategy for skin tissue engineering.
Collapse
Affiliation(s)
- Deyun Chen
- Chinese PLA General Hospital, Beijing, China
| | - Haojie Hao
- Chinese PLA General Hospital, Beijing, China
| | - Chuan Tong
- Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Chinese PLA General Hospital, Beijing, China
| | - Liang Dong
- Chinese PLA General Hospital, Beijing, China
| | - Dongdong Ti
- Chinese PLA General Hospital, Beijing, China
| | - Qian Hou
- Chinese PLA General Hospital, Beijing, China
| | - Huiling Liu
- Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Chinese PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
35
|
Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J Stem Cells 2014; 6:195-202. [PMID: 24772246 PMCID: PMC3999777 DOI: 10.4252/wjsc.v6.i2.195] [Citation(s) in RCA: 276] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/21/2014] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Human umbilical cord (UC) is a promising source of mesenchymal stem cells (MSCs). Apart from their prominent advantages, such as a painless collection procedure and faster self-renewal, UC-MSCs have shown the ability to differentiate into three germ layers, to accumulate in damaged tissue or inflamed regions, to promote tissue repair, and to modulate immune response. There are diverse protocols and culture methods for the isolation of MSCs from the various compartments of UC, such as Wharton’s jelly, vein, arteries, UC lining and subamnion and perivascular regions. In this review, we give a brief introduction to various compartments of UC as a source of MSCs and emphasize the potential clinical utility of UC-MSCs for regenerative medicine and immunotherapy.
Collapse
|
36
|
Gruber HE, Hoelscher GL, Ingram JA, Norton HJ, Hanley EN. Increased IL-17 expression in degenerated human discs and increased production in cultured annulus cells exposed to IL-1ß and TNF-α. Biotech Histochem 2013; 88:302-10. [PMID: 23627571 DOI: 10.3109/10520295.2013.783235] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IL-17 is expressed in a number of tissues including the intervertebral disc, where it exerts strong inflammatory properties. We evaluated IL-17 using immunolocalization in herniated and non-herniated human discs, IL-17 gene expression, and the production of IL-17 by annulus cells cultured in three dimensions in the presence of IL-1ß or TNF-α. There was no difference in the percentage of IL-17 positive cells in annulus or nucleus in herniated vs. non-herniated disc specimens. Molecular studies confirmed expression of IL-17 in disc tissue, with significantly increased expression in more degenerated discs; there was no difference in expression between herniated vs. non-herniated discs. Exposure to IL-1ß or TNF-α resulted in significantly greater production of IL-17. Our findings expand understanding of IL-17 production by disc cells and reveal the importance of non-canonical IL-17 production in the disc. Significantly greater expression of IL-17 in more degenerated discs adds to our understanding of the changes in disc cell function with advancing stages of disc degeneration.
Collapse
Affiliation(s)
- H E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, P.O. Box 32861, Charlotte, NC 28232, USA.
| | | | | | | | | |
Collapse
|
37
|
Zuo Q, Cui W, Liu F, Wang Q, Chen Z, Fan W. Co-cultivated mesenchymal stem cells support chondrocytic differentiation of articular chondrocytes. INTERNATIONAL ORTHOPAEDICS 2013; 37:747-52. [PMID: 23354690 PMCID: PMC3609966 DOI: 10.1007/s00264-013-1782-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 01/07/2013] [Indexed: 11/23/2022]
Abstract
Purpose This study investigated which of the reciprocal stimuli between articular chondrocytes (ACs) and mesenchymal stem cells (MSCs) played the more important role in enhancing cartilage matrix formation, and examined the relative importance of physical contact and soluble factors in the co-culture system. Methods Rat ACs and bone marrow MSCs with green fluorescent protein (GFP-BMSCs) were co-cultured in vitro with or without direct cell–cell contact at the ratio of 2:1. After co-culturing in direct cell–cell contact, ACs and GFP-BMSCs were separated by flow cytometry. The effects of different co-culture methods were analysed by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blotting. Results SOX-9, COL2 and aggrecan mRNA levels and protein expression in ACs co-cultured with direct cell–cell contact were significantly higher than in ACs co-cultured without direct cell–cell contact; and similar results were found in GFP-BMSCs. After co-culture either with or without direct cell–cell contact, mRNA levels and protein expression of SOX-9, COL2 and aggrecan in GFP-BMSCs were significantly lower than in ACs in the equivalent co-culture systems. Though the expression of chondrocyte-specific proteins in GFP-BMSCs was enhanced, the protein expression was still much lower than in ACs cultured alone. Conclusions Reciprocal interactions exist between ACs and BMSCs in co-culture. The stimulating and supporting effects of BMSCs on ACs were more important in enhancing cartilage-matrix formation than the reciprocal effect of ACs on BMSCs. Both soluble factors and direct physical contact occur in AC/BMSC co-cultures, with physical contact playing a predominant, or at least very important role.
Collapse
Affiliation(s)
- Qiang Zuo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | | | | | | | | | | |
Collapse
|
38
|
Peletti-Figueiró M, Silva PGD, Souza OED, Lambert AP, Machado DC, Roesch-Ely M, Henriques JAP, Falavigna A. Stem-cell treatment in disc degeneration: What is the evidence? COLUNA/COLUMNA 2013. [DOI: 10.1590/s1808-18512013000100013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To review the potential role of stem cells in treating degenerative disc disease of the intervertebral disc (IVD). A review was performed of articles from the Medline database concerning stem cells and degenerative disc disease (DDD). To discuss the data, the papers were classified as: review, in vitro, experimental, and clinical. The currently available treatments were basically for symptom reduction, not to revert the IVD degenerative process. The use of mesenchymal stem cells (MSC) is being proposed as an option of treatment for DDD. In vitro studies have shown that the MSC are able to differentiate into NP cells and that the MSC also reduce the inflammatory levels of the degenerated IVD. Besides, experimental studies demonstrated that the MSC remained viable when injected into the IVD, and that they were able to regenerate partially from the degenerated IVD and its structure. The few clinical studies found in the literature presented diverging results. The use of MSC is being widely studied and shows promising results for the treatment of DDD. Although many advances are being achieved in studies in vitro and experimental, there is a lack of clinical studies to prove the role of MSC in DDD management.
Collapse
|
39
|
Gruber HE, Riley FE, Hoelscher GL, Bayoumi EM, Ingram JA, Ramp WK, Bosse MJ, Kellam JF. Osteogenic and chondrogenic potential of biomembrane cells from the PMMA-segmental defect rat model. J Orthop Res 2012; 30:1198-212. [PMID: 22246998 DOI: 10.1002/jor.22047] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 12/05/2011] [Indexed: 02/04/2023]
Abstract
A layer of cells (the "biomembrane") has been identified in large segmental defects between bone and surgically placed methacrylate spacers or antibiotic-impregnated cement beads. We hypothesize that this contains a pluripotent stem cell population with potential valuable applications in orthopedic tissue engineering. Objectives using biomembranes harvested from rat segmental defects were to: (1) Culture biomembrane cells in specialized media to direct progenitor cells along bone or cartilage cell differentiation lineages; (2) evaluate harvested biomembranes for mesenchymal stem cell markers, and (3) define relevant gene expression patterns in harvested biomembranes using microarray analysis. Culture in osteogenic media produced mineralized nodules; culture in chondrogenic media produced masses containing chondroitin sulfate/sulfated proteoglycans. Molecular analysis of biomembrane cells versus control periosteum showed significant upregulation of key genes functioning in mesenchymal stem cell differentiation, development, maintenance, and proliferation. Results identified significant upregulation of WNT receptor signaling pathway genes and significant upregulation of BMP signaling pathway genes. Findings confirm that the biomembrane has a pluripotent stem cell population. The ability to heal large bone defects is clinically challenging, and novel tissue engineering uses of the biomembrane hold great promise in treating non-unions, open fractures with large bone loss and/or infections, and defects associated with tumor resection.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Carolina 28232, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wu L, Prins HJ, Helder MN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells in chondrocyte co-cultures are independent of culture conditions and cell sources. Tissue Eng Part A 2012; 18:1542-51. [PMID: 22429306 DOI: 10.1089/ten.tea.2011.0715] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Earlier, we have shown that the increased cartilage production in pellet co-cultures of chondrocytes and bone marrow-derived mesenchymal stem cells (BM-MSCs) is due to a trophic role of the MSC in stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. These studies were performed in a culture medium that was not compatible with the chondrogenic differentiation of MSCs. In this study, we tested whether the trophic role of the MSCs is dependent on culturing co-culture pellets in a medium that is compatible with the chondrogenic differentiation of MSCs. In addition, we investigated whether the trophic role of the MSCs is dependent on their origins or is a more general characteristic of MSCs. Human BM-MSCs and bovine primary chondrocytes were co-cultured in a medium that was compatible with the chondrogenic differentiation of MSCs. Enhanced matrix production was confirmed by glycosaminoglycans (GAG) quantification. A species-specific quantitative polymerase chain reaction demonstrated that the cartilage matrix was mainly of bovine origin, indicative of a lack of the chondrogenic differentiation of MSCs. In addition, pellet co-cultures were overgrown by bovine cells over time. To test the influence of origin on MSCs' trophic effects, the MSCs isolated from adipose tissue and the synovial membrane were co-cultured with human primary chondrocytes, and their activity was compared with BM-MSCs, which served as control. GAG quantification again confirmed increased cartilage matrix production, irrespective of the source of the MSCs. EdU staining combined with cell tracking revealed an increased proliferation of chondrocytes in each condition. Irrespective of the MSC source, a short tandem repeat analysis of genomic DNA showed a decrease in MSCs in the co-culture over time. Our results clearly demonstrate that in co-culture pellets, the MSCs stimulate cartilage formation due to a trophic effect on the chondrocytes rather than differentiating into chondrocytes, irrespective of culture condition or origin. This implies that the trophic effect of MSCs in co-cultures is a general phenomenon with potential implications for use in cartilage repair strategies.
Collapse
Affiliation(s)
- Ling Wu
- Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
41
|
Gruber HE, Somayaji S, Riley F, Hoelscher GL, Norton HJ, Ingram J, Hanley EN. Human adipose-derived mesenchymal stem cells: serial passaging, doubling time and cell senescence. Biotech Histochem 2012; 87:303-11. [DOI: 10.3109/10520295.2011.649785] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
42
|
Wu L, Leijten JCH, Georgi N, Post JN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng Part A 2011; 17:1425-36. [PMID: 21247341 DOI: 10.1089/ten.tea.2010.0517] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Previous studies showed that coculture of primary chondrocytes (PCs) with various sources of multipotent cells results in a higher relative amount of cartilage matrix formation than cultures containing only chondrocytes. The aim of this study was to investigate the mechanism underlying this observation. We used coculture pellet models of human mesenchymal stem cells (hMSCs) and human PCs or bovine PCs (bPCs) and studied the fate and the contribution to cartilage formation of the individual cell populations during coculture. Enhanced cartilage matrix deposition was confirmed by histology and quantification of total glycosaminoglycan deposition. Species-specific quantitative polymerase chain reaction demonstrated that cartilage matrix gene expression was mainly from bovine origin when bPCs were used. Short tandem repeat analysis and species-specific quantitative polymerase chain reaction analysis of genomic DNA demonstrated the near-complete loss of MSCs in coculture pellets after 4 weeks of culture. In coculture pellets of immortalized MSCs and bPCs, chondrocyte proliferation was increased, which was partly mimicked using conditioned medium, and simultaneously preferential apoptosis of immortalized MSCs was induced. Taken together, our data clearly demonstrate that in pellet cocultures of MSCs and PCs, the former cells disappear over time. Increased cartilage formation in these cocultures is mainly due to a trophic role of the MSCs in stimulating chondrocyte proliferation and matrix deposition by chondrocytes rather than MSCs actively undergoing chondrogenic differentiation.
Collapse
Affiliation(s)
- Ling Wu
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | |
Collapse
|
43
|
Bertolo A, Thiede T, Aebli N, Baur M, Ferguson SJ, Stoyanov JV. Human mesenchymal stem cell co-culture modulates the immunological properties of human intervertebral disc tissue fragments in vitro. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2010; 20:592-603. [PMID: 21181480 DOI: 10.1007/s00586-010-1662-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 11/04/2010] [Accepted: 12/09/2010] [Indexed: 01/08/2023]
Abstract
The capacity of mesenchymal stem cells (MSCs) to differentiate into intervertebral disc (IVD)-like cells has been well described, but their ability to modulate the inflammatory processes in the IVD remains unclear. We found that tissue obtained by discectomy of degenerated and post-traumatic IVD contains significant amounts of IgG antibodies, a sign of lymphocyte infiltration. Further we investigated whether MSCs in vitro, which were characterized for their multilineage differentiation potential and may have immunomodulatory effects on IVD fragments. IVD fragments were co-cultured in contact with peripheral blood lymphocytes (PBLs) and MSCs, and as functional controls we used contact co-cultures of PBLs stimulated with pokeweed mitogen (2.5 μg/mL) and MSCs. The time course of lymphocyte proliferation (Alamar Blue), IgG (ELISA) and gene expression (RT-PCR) of anti-inflammatory cytokines (TGF-β1, IL-10) by MSCs and pro-inflammatory molecules (IL-1α, IL-1β and TNF-α) by the IVD fragments were analyzed. Depending on the response to the presence of MSCs, the IVD fragments (n = 13) were divided in two groups: responders (n = 9), where inflammation was inhibited by MSCs and non-responders (n = 4), where MSCs did not decrease inflammation. At 1 week in co-culture, MSCs reduced significantly the IgG production in the IVD responders group to 69% and PBLs proliferation to 57% of the control. MSCs expression of the anti-inflammatory TGF-β1 increased with time, while IL-10 was expressed only at day 1. IVD gene expression of TNF-α decreased constantly, whereas IL-1α and IL-1β expression increased. In conclusion, these data suggest that MSCs may modulate disc-specific inflammatory and pain status and aid regeneration of the host tissue.
Collapse
|