1
|
Feehan J, Jacques M, Kondrikov D, Eynon N, Wijeratne T, Apostolopoulos V, Gimble JM, Hill WD, Duque G. Circulating Osteoprogenitor Cells Have a Mixed Immune and Mesenchymal Progenitor Function in Humans. Stem Cells 2023; 41:1060-1075. [PMID: 37609930 PMCID: PMC10631805 DOI: 10.1093/stmcls/sxad064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Circulating osteoprogenitors (COP) are a population of cells in the peripheral circulation that possess functional and phenotypical characteristics of multipotent stromal cells (MSCs). This population has a solid potential to become an abundant, accessible, and replenishable source of MSCs with multiple potential clinical applications. However, a comprehensive functional characterization of COP cells is still required to test and fully develop their use in clinical settings. METHODS This study characterized COP cells by comparing them to bone marrow-derived MSCs (BM-MSCs) and adipose-derived MSCs (ASCs) through detailed transcriptomic and proteomic analyses. RESULTS We demonstrate that COP cells have a distinct gene and protein expression pattern with a significantly stronger immune footprint, likely owing to their hematopoietic lineage. In addition, regarding progenitor cell differentiation and proliferation pathways, COP cells have a similar expression pattern to BM-MSCs and ASCs. CONCLUSION COP cells are a unique but functionally similar population to BM-MSCs and ASCs, sharing their proliferation and differentiation capacity, thus presenting an accessible source of MSCs with strong potential for translational regenerative medicine strategies.
Collapse
Affiliation(s)
- Jack Feehan
- Department of Medicine - Western Health, The University of Melbourne, Melbourne, Victoria (VIC), Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Victoria University and University of Melbourne, Melbourne, Victoria (VIC), Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
| | - Macsue Jacques
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, USA
| | - Nir Eynon
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Tissa Wijeratne
- Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Victoria University and University of Melbourne, Melbourne, Victoria (VIC), Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
| | - Vasso Apostolopoulos
- Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Victoria University and University of Melbourne, Melbourne, Victoria (VIC), Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - William D Hill
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, USA
- Department of Veterans Affairs, Ralph H Johnson VA Medical Center, Charleston, SC, USA
| | - Gustavo Duque
- Department of Medicine - Western Health, The University of Melbourne, Melbourne, Victoria (VIC), Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Victoria University and University of Melbourne, Melbourne, Victoria (VIC), Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria (VIC), Australia
- Bone, Muscle and Geroscience Research Group, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Biglari N, Mehdizadeh A, Vafaei Mastanabad M, Gharaeikhezri MH, Gol Mohammad Pour Afrakoti L, Pourbala H, Yousefi M, Soltani-Zangbar MS. Application of mesenchymal stem cells (MSCs) in neurodegenerative disorders: History, findings, and prospective challenges. Pathol Res Pract 2023; 247:154541. [PMID: 37245265 DOI: 10.1016/j.prp.2023.154541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/30/2023]
Abstract
Over the past few decades, the application of mesenchymal stem cells has captured the attention of researchers and practitioners worldwide. These cells can be obtained from practically every tissue in the body and are used to treat a broad variety of conditions, most notably neurological diseases such as Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimer's disease. Studies are still being conducted, and the results of these studies have led to the identification of several different molecular pathways involved in the neuroglial speciation process. These molecular systems are closely regulated and interconnected due to the coordinated efforts of many components that make up the machinery responsible for cell signaling. Within the scope of this study, we compared and contrasted the numerous mesenchymal cell sources and their cellular features. These many sources of mesenchymal cells included adipocyte cells, fetal umbilical cord tissue, and bone marrow. In addition, we investigated whether these cells can potentially treat and modify neurodegenerative illnesses.
Collapse
Affiliation(s)
- Negin Biglari
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Vafaei Mastanabad
- Neurosurgery Department, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | | | - Hooman Pourbala
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
El-Husseiny HM, Kaneda M, Mady EA, Yoshida T, Doghish AS, Tanaka R. Impact of Adipose Tissue Depot Harvesting Site on the Multilineage Induction Capacity of Male Rat Adipose-Derived Mesenchymal Stem Cells: An In Vitro Study. Int J Mol Sci 2023; 24:7513. [PMID: 37108673 PMCID: PMC10138771 DOI: 10.3390/ijms24087513] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Recently, substantial attention has been paid toward adipose-derived mesenchymal stem cells (AdMSCs) as a potential therapy in tissue engineering and regenerative medicine applications. Rat AdMSCs (r-AdMSCs) are frequently utilized. However, the influence of the adipose depot site on the multilineage differentiation potential of the r-AdMSCs is still ambiguous. Hence, the main objective of this study was to explore the influence of the adipose tissue harvesting location on the ability of r-AdMSCs to express the stem-cell-related markers and pluripotency genes, as well as their differentiation capacity, for the first time. Herein, we have isolated r-AdMSCs from the inguinal, epididymal, peri-renal, and back subcutaneous fats. Cells were compared in terms of their phenotype, immunophenotype, and expression of pluripotency genes using RT-PCR. Additionally, we investigated their potential for multilineage (adipogenic, osteogenic, and chondrogenic) induction using special stains confirmed by the expression of the related genes using RT-qPCR. All cells could positively express stem cell marker CD 90 and CD 105 with no significant in-between differences. However, they did not express the hematopoietic markers as CD 34 and CD 45. All cells could be induced successfully. However, epididymal and inguinal cells presented the highest capacity for adipogenic and osteogenic differentiation (21.36-fold and 11.63-fold for OPN, 29.69-fold and 26.68-fold for BMP2, and 37.67-fold and 22.35-fold for BSP, respectively, in epididymal and inguinal cells (p < 0.0001)). On the contrary, the subcutaneous cells exhibited a superior potential for chondrogenesis over the other sites (8.9-fold for CHM1 and 5.93-fold for ACAN, (p < 0.0001)). In conclusion, the adipose tissue harvesting site could influence the differentiation capacity of the isolated AdMSCs. To enhance the results of their employment in various regenerative cell-based therapies, it is thus vital to take the collection site selection into consideration.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Sciences, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Eman A. Mady
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Tadashi Yoshida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11651, Cairo, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| |
Collapse
|
4
|
Deo D, Marchioni M, Rao P. Mesenchymal Stem/Stromal Cells in Organ Transplantation. Pharmaceutics 2022; 14:pharmaceutics14040791. [PMID: 35456625 PMCID: PMC9029865 DOI: 10.3390/pharmaceutics14040791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Organ transplantation is essential and crucial for saving and enhancing the lives of individuals suffering from end-stage organ failure. Major challenges in the medical field include the shortage of organ donors, high rates of organ rejection, and long wait times. To address the current limitations and shortcomings, cellular therapy approaches have been developed using mesenchymal stem/stromal cells (MSC). MSC have been isolated from various sources, have the ability to differentiate to important cell lineages, have anti-inflammatory and immunomodulatory properties, allow immunosuppressive drug minimization, and induce immune tolerance towards the transplanted organ. Additionally, rapid advances in the fields of tissue engineering and regenerative medicine have emerged that focus on either generating new organs and organ sources or maximizing the availability of existing organs. This review gives an overview of the various properties of MSC that have enabled its use as a cellular therapy for organ preservation and transplant. We also highlight emerging fields of tissue engineering and regenerative medicine along with their multiple sub-disciplines, underlining recent advances, widespread clinical applications, and potential impact on the future of tissue and organ transplantation.
Collapse
|
5
|
The miR151 and miR5100 Transfected Bone Marrow Stromal Cells Increase Myoblast Fusion in IGFBP2 Dependent Manner. Stem Cell Rev Rep 2022; 18:2164-2178. [PMID: 35190967 PMCID: PMC9391248 DOI: 10.1007/s12015-022-10350-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
Abstract
Background Bone marrow stromal cells (BMSCs) form a perivascular cell population in the bone marrow. These cells do not present naïve myogenic potential. However, their myogenic identity could be induced experimentally in vitro or in vivo. In vivo, after transplantation into injured muscle, BMSCs rarely fused with myofibers. However, BMSC participation in myofiber reconstruction increased if they were modified by NICD or PAX3 overexpression. Nevertheless, BMSCs paracrine function could play a positive role in skeletal muscle regeneration. Previously, we showed that SDF-1 treatment and coculture with myofibers increased BMSC ability to reconstruct myofibers. We also noticed that SDF-1 treatment changed selected miRNAs expression, including miR151 and miR5100. Methods Mouse BMSCs were transfected with miR151 and miR5100 mimics and their proliferation, myogenic differentiation, and fusion with myoblasts were analyzed. Results We showed that miR151 and miR5100 played an important role in the regulation of BMSC proliferation and migration. Moreover, the presence of miR151 and miR5100 transfected BMSCs in co-cultures with human myoblasts increased their fusion. This effect was achieved in an IGFBP2 dependent manner. Conclusions Mouse BMSCs did not present naïve myogenic potential but secreted proteins could impact myogenic cell differentiation. miR151 and miR5100 transfection changed BMSC migration and IGFBP2 and MMP12 expression in BMSCs. miR151 and miR5100 transfected BMSCs increased myoblast fusion in vitro. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s12015-022-10350-y.
Collapse
|
6
|
A Comparative Study of the Effect of Anatomical Site on Multiple Differentiation of Adipose-Derived Stem Cells in Rats. Cells 2021; 10:cells10092469. [PMID: 34572123 PMCID: PMC8465004 DOI: 10.3390/cells10092469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from adipose tissue are evolved into various cell-based regenerative approaches. Adipose-derived stem cells (ASCs) isolated from rats are commonly used in tissue engineering studies. Still, there is a gap in knowledge about how the harvest locations influence and guide cell differentiation. This study aims to investigate how the harvesting site affects stem-cell-specific surface markers expression, pluripotency, and differentiation potential of ASCs in female Sprague Dawley rats. ASCs were extracted from the adipose tissue of the peri-ovarian, peri-renal, and mesenteric depots and were compared in terms of cell morphology. MSCs phenotype was validated by cell surfaces markers using flow cytometry. Moreover, pluripotent gene expression of Oct4, Nanog, Sox2, Rex-1, and Tert was evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR). ASCs multipotency was evaluated by specific histological stains, and the results were confirmed by quantitative polymerase chain reaction (RT-qPCR) expression analysis of specific genes. There was a non-significant difference detected in the cell morphology and immunophenotype between different harvesting sites. ASCs from multiple locations were significantly varied in their capacity to differentiate into adipocytes, osteoblastic cells, and chondrocytes. To conclude, depot selection is a critical element that should be considered when using ASCs in tissue-specific cell-based regenerative therapies research.
Collapse
|
7
|
Ren L, Guo L, Kou N, Lv J, Wang Z, Yang K. LncRNA LINC00963 promotes osteogenic differentiation of hBMSCs and alleviates osteoporosis progression by targeting miRNA-760/ETS1 axis. Autoimmunity 2021; 54:313-325. [PMID: 34184952 DOI: 10.1080/08916934.2021.1922890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Although long non-coding RNA LINC00963 has been reported to play a crucial regulatory role in osteoporosis (OP), its specific mechanism has not been well studied. Cell viability of human bone marrow mesenchymal stem cells (hBMSCs) transfected with short hairpin RNA targeting LINC00963 (sh-LINC00963) and negative control (sh-NC) was analysed by cell counting kit-8 (CCK-8) assay. Alkaline phosphatase (ALP) activity in hBMSCs transfected with sh-LINC00963 and sh-NC after induction by osteogenic medium (OM) on day 7 was detected. The protein expression levels of osteocalcin (OCN) and osteopontin (OPN) in hBMSCs transfected with sh-LINC00963 and sh-NC during OM induction on day 3 were detected by western blot. The relationship among LINC00963, miR-760, and E26 transformation specific-1 (ETS1) was determined by bioinformatics analysis, luciferase reporter assay, and RNA-binding protein immunoprecipitation (RIP) assay. A rat model with OP was established to confirm the role of LINC00963 in vivo. The expression level of LINC00963 was much lower in hBMSCs isolated from the discarded femoral head tissues of OP patients compared with that in health patients. Meanwhile, the expression level of LINC00963 was significantly increased and the expression level of miR-760 was decreased in hBMSCs during osteogenic induction. LINC00963 could bind to the 3'-untranslated region (3'-UTR) of miR-760 and negatively regulate the expression of miR-760, then promote the osteogenic differentiation in hBMSCs. ETS1 was identified as a target of miR-760. Moreover, overexpression of LINC00963 obviously reduced bone mineral density (BMD) of the left femur in OP rats and alleviated OP progression in vivo. Our results demonstrated that LINC00963 positively regulated the expression of ETS1 by directly targeting miR-760, and then promoted osteogenic differentiation of hBMSCs in vitro, and also attenuated OP progression in vivo, suggesting that LINC00963 might be a potential therapeutic target for OP.
Collapse
Affiliation(s)
- Lirong Ren
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, Dali City, PR China
| | - Limin Guo
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, PR China
| | - Nannan Kou
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, PR China
| | - Jia Lv
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, PR China
| | - Zhihua Wang
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, PR China
| | - Kaishun Yang
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, Dali City, PR China
| |
Collapse
|
8
|
Quality by design to define critical process parameters for mesenchymal stem cell expansion. Biotechnol Adv 2021; 50:107765. [PMID: 33961977 DOI: 10.1016/j.biotechadv.2021.107765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/01/2021] [Indexed: 12/15/2022]
Abstract
Stem cell-based therapeutic products could be the key to treat the deadliest current pathologies, ranging from neuro-degenerative to respiratory diseases. However, in order to bring these innovative therapeutics to a commercialization stage, reproducible manufacturing of high quality cell products is required. Although advances in cell culture techniques have led to more robust production processes and dramatically accelerated the development of early-phase clinical studies, challenges remain before regulatory approval, particularly to define and implement science-based quality standards (essential pre-requisites for national health agencies). In this regard, using new methodologies, such as Quality By Design (QBD), to build the production process around drug quality, could significantly reduce the chance of product rejection. This review-based work aims to perform a QBD approach to Mesenchymal Stem Cell (MSC) manufacturing in standard two-dimensional flasks, using published studies which have determined the impact of individual process parameters on defined Critical Quality Attributes (CQA). Along with this bibliographic analysis, parameter criticality was determined during the two main manufacturing stages (cell extraction and cell amplification) along with an overall classification in view of identifying the Critical Process Parameters (CPP). The analysis was performed in view of an improved standardization between research teams, and should contribute to reduce the gap towards compliant Good Manufacturing Practice (cGMP) manufacturing.
Collapse
|
9
|
Integrated analysis of miRNA and mRNA transcriptomic reveals antler growth regulatory network. Mol Genet Genomics 2021; 296:689-703. [PMID: 33770271 DOI: 10.1007/s00438-021-01776-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/15/2021] [Indexed: 01/26/2023]
Abstract
The growth of antler is driven by endochondral ossification in the growth center of the apical region. Antler grows faster than cancer tissues, but it can be stably regulated and regenerated periodically. To elucidate the molecular mechanisms of how antler grows rapidly without carcinogenesis, in this study, we used RNA-seq technology to evaluate the changes of miRNA and mRNA profiles in antler at four different developmental stages, including 15, 60, 90, and 110 days. We identified a total of 55004 unigenes and 246 miRNAs of which, 10182, 13258, 10740 differentially expressed (DE) unigenes and 35, 53, 27 DE miRNAs were identified in 60-day vs. 15-day, 90-day vs. 60-day, and 110-day vs. 90-day. GO and KEGG pathway analysis indicated that DE unigenes and DE miRNA were mainly associated with chondrogenesis, osteogenesis and inhibition of oncogenesis, that were closely related to antler growth. The interaction networks of mRNA-mRNA and miRNA-mRNA related to chondrogenesis, osteogenesis and inhibition of oncogenesis of antler were constructed. The results indicated that mRNAs (COL2A1, SOX9, WWP2, FGFR1, SPARC, LOX, etc.) and miRNAs (miR-145, miR-199a-3p, miR-140, miR-199a-5p, etc.) might have key roles in chondrogenesis and osteogenesis of antler. As well as mRNA (TP53, Tpm3 and ATP1A1, etc.) and miRNA (miR-106a, miR-145, miR-1260b and miR-2898, etc.) might play important roles in inhibiting the carcinogenesis of antler. In summary, we constructed the mRNA-mRNA and miRNA-mRNA regulatory networks related to chondrogenesis, osteogenesis and inhibition of oncogenesis of antler, and identified key candidate mRNAs and miRNAs among them. Further developments and validations may provide a reference for in-depth analysis of the molecular mechanism of antler growth without carcinogenesis.
Collapse
|
10
|
Jorgensen C, Khoury M. Musculoskeletal Progenitor/Stromal Cell-Derived Mitochondria Modulate Cell Differentiation and Therapeutical Function. Front Immunol 2021; 12:606781. [PMID: 33763061 PMCID: PMC7982675 DOI: 10.3389/fimmu.2021.606781] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
Musculoskeletal stromal cells’ (MSCs’) metabolism impacts cell differentiation as well as immune function. During osteogenic and adipogenic differentiation, BM-MSCs show a preference for glycolysis during proliferation but shift to an oxidative phosphorylation (OxPhos)-dependent metabolism. The MSC immunoregulatory fate is achieved with cell polarization, and the result is sustained production of immunoregulatory molecules (including PGE2, HGF, IL1RA, IL6, IL8, IDO activity) in response to inflammatory stimuli. MSCs adapt their energetic metabolism when acquiring immunomodulatory property and shift to aerobic glycolysis. This can be achieved via hypoxia, pretreatment with small molecule-metabolic mediators such as oligomycin, or AKT/mTOR pathway modulation. The immunoregulatory effect of MSC on macrophages polarization and Th17 switch is related to the glycolytic status of the MSC. Indeed, MSCs pretreated with oligomycin decreased the M1/M2 ratio, inhibited T-CD4 proliferation, and prevented Th17 switch. Mitochondrial activity also impacts MSC metabolism. In the bone marrow, MSCs are present in a quiescent, low proliferation, but they keep their multi-progenitor function. In this stage, they appear to be glycolytic with active mitochondria (MT) status. During MSC expansion, we observed a metabolic shift toward OXPhos, coupled with an increased MT activity. An increased production of ROS and dysfunctional mitochondria is associated with the metabolic shift to glycolysis. In contrast, when MSC underwent chondro or osteoblast differentiation, they showed a decreased glycolysis and inhibition of the pentose phosphate pathway (PPP). In parallel the mitochondrial enzymatic activities increased associated with oxidative phosphorylation enhancement. MSCs respond to damaged or inflamed tissue through the transfer of MT to injured and immune cells, conveying a type of signaling that contributes to the restoration of cell homeostasis and immune function. The delivery of MT into injured cells increased ATP levels which in turn maintained cellular bioenergetics and recovered cell functions. MSC-derived MT may be transferred via tunneling nanotubes to undifferentiated cardiomyocytes and leading to their maturation. In this review, we will decipher the pathways and the mechanisms responsible for mitochondria transfer and activity. The eventual reversal of the metabolic and pro-inflammatory profile induced by the MT transfer will open new avenues for the control of inflammatory diseases.
Collapse
Affiliation(s)
- Christian Jorgensen
- Inserm, U1183, Montpellier, France.,Université MONTPELLIER 1, UFR de Médecine, Montpellier, France.,Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, France
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
11
|
Ai L, Yi W, Chen L, Wang H, Huang Q. Xian-Ling-Gu-Bao protects osteoporosis through promoting osteoblast differentiation by targeting miR-100-5p/KDM6B/RUNX2 axis. In Vitro Cell Dev Biol Anim 2021; 57:3-9. [PMID: 33398630 DOI: 10.1007/s11626-020-00530-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/10/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Liang Ai
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.,Department of Traditional Chinese Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Weimin Yi
- Department of Traditional Chinese Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Liudan Chen
- Department of Traditional Chinese Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Haibin Wang
- Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Qihui Huang
- Department of Traditional Chinese Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China.
| |
Collapse
|
12
|
Gugjoo MB, Hussain S, Amarpal, Shah RA, Dhama K. Mesenchymal Stem Cell-Mediated Immuno-Modulatory and Anti- Inflammatory Mechanisms in Immune and Allergic Disorders. ACTA ACUST UNITED AC 2020; 14:3-14. [PMID: 32000656 PMCID: PMC7509741 DOI: 10.2174/1872213x14666200130100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 01/25/2020] [Accepted: 01/25/2020] [Indexed: 02/06/2023]
Abstract
Background: Mesenchymal Stem Cells (MSCs) are present in almost all the tissues of the body and act as the backbone of the internal tissue homeostasis. Among their various characteristic features, immuno-modulatory and/ anti-inflammatory properties play an important role in therapeutics. Objective: The current topic focuses on the characterization and immuno-modulatory and/ anti-inflammatory properties of MSCs. To present and discuss the current status of MSCs immuno-modulatory properties. Methods: Available literature on MSCs properties and patents have been detailed, critically interpreted, and discussed based upon available literature. The main focus has been on their characteristic immuno-modulatory and anti-inflammatory properties though some of the basic characterization markers have also been detailed. The databases searched for the literature include PubMed, Med Line, PubMed Central, Science Direct and a few other scientific databases. Results: MSCs are present in a very limited concentration in the tissues, and as such their culture expansion becomes imperative. MSCs immuno-modulatory and anti-inflammatory roles are achieved through direct cell-cell contact and / by the release of certain factors. Such properties are controlled by micro-environment upon which currently very limited control can be exerted. Besides, further insights in the xeno-protein free culture media as against the fetal bovine serum is required. Conclusion: MSCs have been well-isolated, cultured and characterized from numerous tissues of the body. The majority of the studies have shown MSCs as immuno-compromised with immunomodulatory and / or anti-inflammatory properties except some of the latest studies that have failed to achieve the desired results and thus, demand further research. Further research is required in the area to translate the results into clinical application.
Collapse
Affiliation(s)
- Mudasir B Gugjoo
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST, Shuhama, Srinagar-190006, Jammu and Kashmir, India
| | - Shahid Hussain
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST, Shuhama, Srinagar-190006, Jammu and Kashmir, India
| | - Amarpal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243 122, Uttar Pradesh, India
| | - Riaz A Shah
- Divison of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST, Shuhama, Srinagar-190006, Jammu and Kashmir, India
| | - Kuldeep Dhama
- Division of Pathology, ICARIndian Veterinary Research Institute, Izatnagar, Bareilly, 243 122, Uttar Pradesh, India
| |
Collapse
|
13
|
Sharifi S, Moghaddam FA, Abedi A, Maleki Dizaj S, Ahmadian S, Abdolahinia ED, Khatibi SMH, Samiei M. Phytochemicals impact on osteogenic differentiation of mesenchymal stem cells. Biofactors 2020; 46:874-893. [PMID: 33037744 DOI: 10.1002/biof.1682] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Medicinal plants have always been utilized for the prevention and treatment of the spread of different diseases all around the world. To name some traditional medicine that has been used over centuries, we can refer to phytochemicals such as naringin, icariin, genistein, and resveratrol gained from plants. Osteogenic differentiation and mineralization of stem cells can be the result of specific bioactive compounds from plants. One of the most appealing choices for therapy can be mesenchymal stem cells (MSCs) because it has a great capability of self-renewal and differentiation into three descendants, namely, endoderm, mesoderm, and ectoderm. Stem cell gives us the glad tidings of great advances in tissue regeneration and transplantation field for treatment of diseases. Using plant bioactive phytochemicals also holds tremendous promises in treating diseases such as osteoporosis. The purpose of the present review article thus is to investigate what are the roles and consequences of phytochemicals on osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Atefeh Abedi
- Department of Endodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahin Ahmadian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center of Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Samiei
- Department of Endodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Lu M, Guo J, Wu B, Zhou Y, Wu M, Farzaneh M, Khoshnam SE. Mesenchymal Stem Cell-Mediated Mitochondrial Transfer: a Therapeutic Approach for Ischemic Stroke. Transl Stroke Res 2020; 12:212-229. [PMID: 32975692 DOI: 10.1007/s12975-020-00853-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022]
Abstract
Stroke is the leading cause of death and adult disability worldwide. Mitochondrial dysfunction is one of the hallmarks of stroke-induced neuronal death, and maintaining mitochondrial function is essential in cell survival and neurological progress following ischemic stroke. Stem cell-mediated mitochondrial transfer represents an emerging therapeutic approach for ischemic stroke. Accumulating evidence suggests that mesenchymal stem cells (MSCs) can directly transfer healthy mitochondria to damaged cells, and rescue mitochondrial damage-provoked tissue degeneration. This review summarizes the research on MSCs-mediated mitochondrial transfer as a therapeutic strategy against ischemic stroke.
Collapse
Affiliation(s)
- Meng Lu
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Jindong Guo
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Bowen Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Biochemistry, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yuhui Zhou
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Mishan Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China. .,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
15
|
Fan XL, Zhang Y, Li X, Fu QL. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol Life Sci 2020; 77:2771-2794. [PMID: 31965214 PMCID: PMC7223321 DOI: 10.1007/s00018-020-03454-6] [Citation(s) in RCA: 299] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) have been extensively investigated for the treatment of various diseases. The therapeutic potential of MSCs is attributed to complex cellular and molecular mechanisms of action including differentiation into multiple cell lineages and regulation of immune responses via immunomodulation. The plasticity of MSCs in immunomodulation allow these cells to exert different immune effects depending on different diseases. Understanding the biology of MSCs and their role in treatment is critical to determine their potential for various therapeutic applications and for the development of MSC-based regenerative medicine. This review summarizes the recent progress of particular mechanisms underlying the tissue regenerative properties and immunomodulatory effects of MSCs. We focused on discussing the functional roles of paracrine activities, direct cell-cell contact, mitochondrial transfer, and extracellular vesicles related to MSC-mediated effects on immune cell responses, cell survival, and regeneration. This will provide an overview of the current research on the rapid development of MSC-based therapies.
Collapse
Affiliation(s)
- Xing-Liang Fan
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road II, Guangzhou, 510080, People's Republic of China
| | - Yuelin Zhang
- Department of Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Road II, Guangzhou, 510080, People's Republic of China
| | - Xin Li
- Department of Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Road II, Guangzhou, 510080, People's Republic of China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road II, Guangzhou, 510080, People's Republic of China.
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
16
|
Rambøl MH, Han E, Niklason LE. Microvessel Network Formation and Interactions with Pancreatic Islets in Three-Dimensional Chip Cultures. Tissue Eng Part A 2020; 26:556-568. [PMID: 31724494 PMCID: PMC7249478 DOI: 10.1089/ten.tea.2019.0186] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
The pancreatic islet is a highly vascularized micro-organ, and rapid revascularization postislet transplantation is important for islet survival and function. However, the various mechanisms involved in islet revascularization are not fully understood, and we currently lack good in vitro platforms to explore this. Our aim for this study was to generate perfusable microvascular networks in a microfluidic chip device, in which islets could be easily integrated, to establish an in vitro platform for investigations on islet-microvasculature interactions. We compared the ability of mesenchymal stem cells (MSCs) and fibroblasts to support microvascular network formation by human umbilical vein endothelial cells (HUVECs) and human induced pluripotent stem cell-derived endothelial colony-forming cell in two-dimensional and three-dimensional models of angiogenesis, and tested the effect of different culture media on microvessel formation. HUVECs that were supported by MSCs formed patent and perfusable networks in a fibrin gel, whereas networks supported by fibroblasts rapidly regressed. Network morphology could be controlled by adjusting relative cell numbers and densities. Incorporation of isolated rat islets demonstrated that islets recruit local microvasculature in vitro, but that the microvessels did not invade islets, at least during the course of these studies. This in vitro microvascularization platform can provide a useful tool to study how various parameters affect islet integration with microvascular networks and could also be utilized for studies of vascularization of other organ systems. Impact statement To improve pancreatic islet graft survival and function posttransplantation, rapid and adequate revascularization is critical. Efforts to improve islet revascularization are demanding due to an insufficient understanding of the mechanisms involved in the process. We have applied a microfluidics platform to generate microvascular networks, and by incorporating pancreatic islets, we were able to study microvasculature-islet interactions in real time. This platform can provide a useful tool to study islet integration with microvascular networks, and could be utilized for studies of vascularization of other organ systems. Moreover, this work may be adapted toward developing a prevascularized islet construct for transplantation.
Collapse
Affiliation(s)
- Mia H. Rambøl
- Department of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Edward Han
- Department of Biomedical Engineering and Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| | - Laura E. Niklason
- Department of Biomedical Engineering and Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Baptista LS. Adipose stromal/stem cells in regenerative medicine: Potentials and limitations. World J Stem Cells 2020; 12:1-7. [PMID: 32110271 PMCID: PMC7031762 DOI: 10.4252/wjsc.v12.i1.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
This article presents the stem and progenitor cells from subcutaneous adipose tissue, briefly comparing them with their bone marrow counterparts, and discussing their potential for use in regenerative medicine. Subcutaneous adipose tissue differs from other mesenchymal stromal/stem cells (MSCs) sources in that it contains a pre-adipocyte population that dwells in the adventitia of robust blood vessels. Pre-adipocytes are present both in the stromal-vascular fraction (SVF; freshly isolated cells) and in the adherent fraction of adipose stromal/stem cells (ASCs; in vitro expanded cells), and have an active role on the chronic inflammation environment established in obesity, likely due their monocytic-macrophage lineage identity. The SVF and ASCs have been explored in cell therapy protocols with relative success, given their paracrine and immunomodulatory effects. Importantly, the widely explored multipotentiality of ASCs has direct application in bone, cartilage and adipose tissue engineering. The aim of this editorial is to reinforce the peculiarities of the stem and progenitor cells from subcutaneous adipose tissue, revealing the spheroids as a recently described biotechnological tool for cell therapy and tissue engineering. Innovative cell culture techniques, in particular 3D scaffold-free cultures such as spheroids, are now available to increase the potential for regeneration and differentiation of mesenchymal lineages. Spheroids are being explored not only as a model for cell differentiation, but also as powerful 3D cell culture tools to maintain the stemness and expand the regenerative and differentiation capacities of mesenchymal cell lineages.
Collapse
Affiliation(s)
- Leandra Santos Baptista
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| |
Collapse
|
18
|
Mantripragada VP, Bova WA, Piuzzi NS, Boehm C, Obuchowski NA, Midura RJ, Muschler GF. Native-Osteoarthritic Joint Resident Stem and Progenitor Cells for Cartilage Cell-Based Therapies: A Quantitative Comparison With Respect to Concentration and Biological Performance. Am J Sports Med 2019; 47:3521-3530. [PMID: 31671273 DOI: 10.1177/0363546519880905] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cell-based therapy for cartilage repair is a promising approach and is becoming an established technique. Yet, there is no consensus on the optimal cell source. PURPOSE To provide a donor-matched quantitative comparison of the connective tissue progenitors (CTPs) derived from cartilage (Outerbridge grade 1-3 [G1-2-3]), bone marrow aspirate concentrate (BMC), infrapatellar fat pad (IPFP), synovium, and periosteum with respect to (1) cell concentration ([Cell], cells/mL), (2) CTP prevalence (PCTP, colonies per million cells), and (3) biological performance based on in vitro proliferation potential (cells per colony) colony density, and differentiation potential (expression of negatively charged extracellular matrix: glycosaminoglycan-rich extra cellular matrix [GAG-ECM]). STUDY DESIGN Descriptive laboratory study. METHODS Tissues were obtained from 10 patients undergoing total knee arthroplasty (mean age, 59 years; women, n = 6). Automated quantitative colony-forming unit analysis was used to compare [Cell], PCTP, and CTP biological performance across tissue sources. RESULTS [Cell] was highest in grade 3 cartilage (P = .002) and BMC (P = .001). Median PCTP was highest in IPFP (P = .001), synovium (P = .003), and G1-2 cartilage (P = .02). Proliferation was highest in synovium-derived CTPs (P < .001). Median colony density was highest in G1-2-3 (P < .001). Median GAG-ECM was highest in G1-2-3 (P < .001). Within each patient, CTPs derived from all tissues were highly heterogeneous in biological performance as determined by cells per colony, density, and GAG-ECM. CONCLUSION Tissue sources differ in [Cell], PCTP, and biological attributes. The data presented in this study suggest that cartilage (G1-2-3) is the preferred tissue source for cartilage repair based on PCTP and GAG-ECM, followed by synovium, IPFP, BMC, and periosteum. However, due to the heterogeneous mixture of CTPs within each tissue source, there exists a subset of CTPs with biological performance similar to G1-2-3 cartilage, particularly in synovium and IPFP. Performance-based clonal selection and expansion of preferred CTPs and their progeny will potentially lead to improved cell population with predictive future. CLINICAL RELEVANCE Optimal tissue regeneration strategies will require informed decisions regarding which of the available tissue sources to use. Optimizing cell sourcing in any tissue may require separation of CTPs with preferred attributes from those with less desirable attributes. The heterogeneity manifest in the early stage of colony formation represents an opportunity for performance-based clone selection for clinical cell processing and manufacturing.
Collapse
Affiliation(s)
- Venkata P Mantripragada
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wes A Bova
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicolas S Piuzzi
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cynthia Boehm
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nancy A Obuchowski
- Department of Quantitative Health Science, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ronald J Midura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - George F Muschler
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
19
|
Saud B, Malla R, Shrestha K. A Review on the Effect of Plant Extract on Mesenchymal Stem Cell Proliferation and Differentiation. Stem Cells Int 2019; 2019:7513404. [PMID: 31428160 PMCID: PMC6681598 DOI: 10.1155/2019/7513404] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/29/2019] [Indexed: 02/07/2023] Open
Abstract
Stem cell has immense potential in regenerative cellular therapy. Mesenchymal stem cells (MSCs) can become a potential attractive candidate for therapy due to its remarkable ability of self-renewal and differentiation into three lineages, i.e., ectoderm, mesoderm, and endoderm. Stem cell holds tremendous promises in the field of tissue regeneration and transplantation for disease treatments. Globally, medicinal plants are being used for the treatment and prevention of a variety of diseases. Phytochemicals like naringin, icariin, genistein, and resveratrol obtained from plants have been extensively used in traditional medicine for centuries. Certain bioactive compounds from plants increase the rate of tissue regeneration, differentiation, and immunomodulation. Several studies show that bioactive compounds from plants have a specific role (bioactive mediator) in regulating the rate of cell division and differentiation through complex signal pathways like BMP2, Runx2, and Wnt. The use of plant bioactive phytochemicals may also become promising in treating diseases like osteoporosis, neurodegenerative disorders, and other tissue degenerative disorders. Thus, the present review article is aimed at highlighting the roles and consequences of plant extracts on MSCs proliferation and desired lineage differentiations.
Collapse
Affiliation(s)
- Bhuvan Saud
- Central Department of Biotechnology, Tribhuvan University, Kirtipur, Nepal
- Faculty of Science, Nepal Academy of Science and Technology (NAST), Khumaltar, Lalitpur, Nepal
| | - Rajani Malla
- Central Department of Biotechnology, Tribhuvan University, Kirtipur, Nepal
| | - Kanti Shrestha
- Faculty of Science, Nepal Academy of Science and Technology (NAST), Khumaltar, Lalitpur, Nepal
| |
Collapse
|
20
|
Kim W, Lee SK, Kwon YW, Chung SG, Kim S. Pioglitazone-Primed Mesenchymal Stem Cells Stimulate Cell Proliferation, Collagen Synthesis and Matrix Gene Expression in Tenocytes. Int J Mol Sci 2019; 20:ijms20030472. [PMID: 30678291 PMCID: PMC6387004 DOI: 10.3390/ijms20030472] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 11/16/2022] Open
Abstract
Various therapeutic effects of mesenchymal stem cells (MSCs) have been reported. However, the rapid clearance of these cells in vivo, difficulties in identifying their therapeutic mechanism of action, and insufficient production levels remain to be resolved. We investigated whether a pioglitazone pre-treatment of MSCs (Pio-MSCs) would stimulate the proliferation of co-cultured tenocytes. Pioglitazone increased the proliferation of MSCs and enhanced the secretion of VEGF (vascular endothelial growth factor) and collagen in these cells. We then examined the effects of Pio-MSCs on tenocytes using an indirect transwell culture system. A significant increase in tenocyte proliferation and cell cycle progression was observed in these co-cultures. Significant increases were observed in wound scratch closure by tenocytes from a Pio-MSC co-culture. Pio-MSCs also enhanced the secretion of collagen from tenocytes. A higher mRNA level of collagen type 1 (Col 1) and type 3 (Col 3), scleraxis (Scx), and tenascin C (TnC) was found in the tenocytes in Pio-MSC co-cultures compared with monocultured cells or tenocytes cultured with non-treated MSCs. Our results indicate that pioglitazone enhances the therapeutic effects of MSCs on tendon repair.
Collapse
Affiliation(s)
- Won Kim
- Department of Rehabilitation Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
- Department of Rehabilitation Medicine, College of Medicine, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
| | - Seul Ki Lee
- Stem Cell Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea.
| | - Young-Won Kwon
- Department of Rehabilitation Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Sun G Chung
- Department of Rehabilitation Medicine, College of Medicine, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
- Institute of Aging, Seoul National University, Seoul 03080, Korea.
- Rheumatism Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea.
| | - Soo Kim
- Stem Cell Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea.
| |
Collapse
|
21
|
Mesenchymal Stromal Cells and Cutaneous Wound Healing: A Comprehensive Review of the Background, Role, and Therapeutic Potential. Stem Cells Int 2018; 2018:6901983. [PMID: 29887893 PMCID: PMC5985130 DOI: 10.1155/2018/6901983] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
Cutaneous wound repair is a highly coordinated cascade of cellular responses to injury which restores the epidermal integrity and its barrier functions. Even under optimal healing conditions, normal wound repair of adult human skin is imperfect and delayed healing and scarring are frequent occurrences. Dysregulated wound healing is a major concern for global healthcare, and, given the rise in diabetic and aging populations, this medicoeconomic disease burden will continue to rise. Therapies to reliably improve nonhealing wounds and reduce scarring are currently unavailable. Mesenchymal stromal cells (MSCs) have emerged as a powerful technique to improve skin wound healing. Their differentiation potential, ease of harvest, low immunogenicity, and integral role in native wound healing physiology make MSCs an attractive therapeutic remedy. MSCs promote cell migration, angiogenesis, epithelialization, and granulation tissue formation, which result in accelerated wound closure. MSCs encourage a regenerative, rather than fibrotic, wound healing microenvironment. Recent translational research efforts using modern bioengineering approaches have made progress in creating novel techniques for stromal cell delivery into healing wounds. This paper discusses experimental applications of various stromal cells to promote wound healing and discusses the novel methods used to increase MSC delivery and efficacy.
Collapse
|
22
|
Arima Y, Hokimoto S, Tabata N, Nakagawa O, Oshima A, Matsumoto Y, Sato T, Mukunoki T, Otani J, Ishii M, Uchikawa M, Yamamoto E, Izumiya Y, Kaikita K, Ogawa H, Nishiyama K, Tsujita K. Evaluation of Collateral Source Characteristics With 3-Dimensional Analysis Using Micro-X-Ray Computed Tomography. J Am Heart Assoc 2018; 7:JAHA.117.007800. [PMID: 29572323 PMCID: PMC5907550 DOI: 10.1161/jaha.117.007800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Collateral arteries provide an alternative blood supply and protect tissues from ischemic damage in patients with peripheral artery disease. However, the mechanism of collateral artery development is difficult to validate. METHODS AND RESULTS Collateral arteries were visualized using micro-x-ray computed tomography. Developmental characteristics were assessed using confocal microscopy. We conducted a single-center, retrospective, observational study and assessed the dilatation of collateral arteries on ischemic sides. We quantified the vascular volume in both ischemic and nonischemic legs. A prominent increase in vascular volume was observed in the ischemic leg using a murine hind-limb ischemia model. We also performed qualitative assessment and confirmed that the inferior gluteal artery functioned as a major collateral source. Serial analysis of murine hind-limb vessel development revealed that the inferior gluteal artery was a remnant of the ischial artery, which emerged as a representative vessel on the dorsal side during hind-limb organogenesis. We retrospectively analyzed consecutive patients who were admitted for the diagnosis or treatment of peripheral artery disease. The diameter of the inferior gluteal artery on the ischemic side showed significant dilatation compared with that on the nonischemic side. CONCLUSIONS Our findings indicate that an embryonic remnant artery can become a collateral source under ischemic conditions. Flow enhancement in the inferior gluteal artery might become a novel therapeutic approach for patients with peripheral artery disease.
Collapse
Affiliation(s)
- Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan .,X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto, Japan.,Department of Molecular Physiology, National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan.,Kyushu University of Nursing and Social Welfare, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Asahi Oshima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Matsumoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Sato
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Toshifumi Mukunoki
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Jun Otani
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Michie Uchikawa
- International Research Center for Medical Sciences, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisao Ogawa
- National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Koichi Nishiyama
- International Research Center for Medical Sciences, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
23
|
Azizi Z, Lange C, Paroni F, Ardestani A, Meyer A, Wu Y, Zander AR, Westenfelder C, Maedler K. β-MSCs: successful fusion of MSCs with β-cells results in a β-cell like phenotype. Oncotarget 2018; 7:48963-48977. [PMID: 27374092 PMCID: PMC5226484 DOI: 10.18632/oncotarget.10214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 04/19/2016] [Indexed: 12/22/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (MSC) have anti-inflammatory, anti-apoptotic and immunosuppressive properties and are a potent source for cell therapy. Cell fusion has been proposed for rapid generation of functional new reprogrammed cells. In this study, we aimed to establish a fusion protocol of bone marrow−derived human MSCs with the rat beta-cell line (INS-1E) as well as human isolated pancreatic islets in order to generate insulin producing beta-MSCs as a cell-based treatment for diabetes. Human eGFP+ puromycin+ MSCs were co-cultured with either stably mCherry-expressing rat INS-1E cells or human dispersed islet cells and treated with phytohemagglutinin (PHA-P) and polyethylene glycol (PEG) to induce fusion. MSCs and fused cells were selected by puromycin treatment. With an improved fusion protocol, 29.8 ± 2.9% of all MSCs were β-MSC heterokaryons based on double positivity for mCherry and eGFP. After fusion and puromycin selection, human NKX6.1 and insulin as well as rat Neurod1, Nkx2.2, MafA, Pdx1 and Ins1 mRNA were highly elevated in fused human MSC/INS-1E cells, compared to the mixed control population. Such induction of beta-cell markers was confirmed in fused human MSC/human dispersed islet cells, which showed elevated NEUROD1, NKX2.2, MAFA, PDX1 and insulin mRNA compared to the mixed control. Fused cells had higher insulin content and improved insulin secretion compared to the mixed control and insulin positive beta-MSCs also expressed nuclear PDX1. We established a protocol for fusion of human MSCs and beta cells, which resulted in a beta cell like phenotype. This could be a novel tool for cell-based therapies of diabetes.
Collapse
Affiliation(s)
- Zahra Azizi
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Lange
- Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Federico Paroni
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Amin Ardestani
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Anke Meyer
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Yonghua Wu
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Axel R Zander
- Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christof Westenfelder
- Departments of Medicine and Physiology, University of Utah and George E Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Kathrin Maedler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,German Center for Diabetes Research (DZD) project partner, University of Bremen, Bremen, Germany
| |
Collapse
|
24
|
Adipose Stem Cells: From Bench to Bedside. Stem Cells Int 2016; 2016:6484038. [PMID: 27087816 PMCID: PMC4819109 DOI: 10.1155/2016/6484038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 02/28/2016] [Indexed: 12/26/2022] Open
|
25
|
Wang H, Zhou Y, Chu TW, Li CQ, Wang J, Zhang ZF, Huang B. Distinguishing characteristics of stem cells derived from different anatomical regions of human degenerated intervertebral discs. 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 2016; 25:2691-704. [PMID: 26984881 DOI: 10.1007/s00586-016-4522-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/05/2016] [Accepted: 03/06/2016] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Several types of stem cells have been successfully demonstrated to exist in the human degenerated intervertebral disc (IVD), which is composed of annulus fibrosus (AF), nucleus pulposus (NP) and cartilage endplate (CEP). However, the differences in the biological characteristics among these and bone marrow derived mesenchymal stem cells (BM-MSCs) remain unclear. MATERIALS AND METHODS To investigate this issue, cells were harvested from human AF, NP, CEP, and bone marrow, respectively; passage 2 cells were selected using the agarose suspension culture system to obtain stem cell clones. Following expansion in vitro, stem cells from different anatomical regions were compared regarding the morphology, proliferation ability, immunophenotypic expression, and multi-lineage differentiation capacity. In addition, stem cell-alginate bead compositions were constructed for the comparison of DNA and sGAG content. RESULTS There were subtle differences regarding cell morphology, but no significant differences in proliferation ability among the four types of stem cells. For the immunophenotypic analysis, all stem cells basically fulfilled the criteria for mesenchymal stem cells (MSCs), which have been published by the International Society for Cellular Therapy (ISCT), with a significant difference in CD105 expression. A comparison of the osteogenic capacities indicated: cartilage endplate-derived stem cells (CESCs) > annulus fibrosus-derived stem cells (AFSCs) > BM-MSCs > nucleus pulposus-derived stem cells (NPSCs). The chondrogenesis difference was similar to osteogenesis. For adipogenesis: BM-MSCs >NPSCs >CESCs >AFSCs. In the stem cell/alginate composition, the CESCs consistently showed the superior chondrogenic potential among all those cell types. CONCLUSIONS Our data indicated that all the four types of stem cells shared some similar biological properties (regarding shape, proliferation ability and immunophenotypic expression). CESCs, which had the strongest osteogenic and chondrogenic potentials, may serve as excellent seed cells for NP/cartilage or bone tissue engineering.
Collapse
Affiliation(s)
- Hai Wang
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, People's Republic of China
| | - Yue Zhou
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Tong-Wei Chu
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Chang-Qing Li
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Jian Wang
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Zheng-Feng Zhang
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Bo Huang
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
26
|
Ross CL, Siriwardane M, Almeida-Porada G, Porada CD, Brink P, Christ GJ, Harrison BS. The effect of low-frequency electromagnetic field on human bone marrow stem/progenitor cell differentiation. Stem Cell Res 2015; 15:96-108. [PMID: 26042793 PMCID: PMC4516580 DOI: 10.1016/j.scr.2015.04.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/17/2015] [Accepted: 04/27/2015] [Indexed: 12/12/2022] Open
Abstract
Human bone marrow stromal cells (hBMSCs, also known as bone marrow-derived mesenchymal stem cells) are a population of progenitor cells that contain a subset of skeletal stem cells (hSSCs), able to recreate cartilage, bone, stroma that supports hematopoiesis and marrow adipocytes. As such, they have become an important resource in developing strategies for regenerative medicine and tissue engineering due to their self-renewal and differentiation capabilities. The differentiation of SSCs/BMSCs is dependent on exposure to biophysical and biochemical stimuli that favor early and rapid activation of the in vivo tissue repair process. Exposure to exogenous stimuli such as an electromagnetic field (EMF) can promote differentiation of SSCs/BMSCs via ion dynamics and small signaling molecules. The plasma membrane is often considered to be the main target for EMF signals and most results point to an effect on the rate of ion or ligand binding due to a receptor site acting as a modulator of signaling cascades. Ion fluxes are closely involved in differentiation control as stem cells move and grow in specific directions to form tissues and organs. EMF affects numerous biological functions such as gene expression, cell fate, and cell differentiation, but will only induce these effects within a certain range of low frequencies as well as low amplitudes. EMF has been reported to be effective in the enhancement of osteogenesis and chondrogenesis of hSSCs/BMSCs with no documented negative effects. Studies show specific EMF frequencies enhance hSSC/BMSC adherence, proliferation, differentiation, and viability, all of which play a key role in the use of hSSCs/BMSCs for tissue engineering. While many EMF studies report significant enhancement of the differentiation process, results differ depending on the experimental and environmental conditions. Here we review how specific EMF parameters (frequency, intensity, and time of exposure) significantly regulate hSSC/BMSC differentiation in vitro. We discuss optimal conditions and parameters for effective hSSC/BMSC differentiation using EMF treatment in an in vivo setting, and how these can be translated to clinical trials.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest Institute for Regenerative Medicine, USA; Wake Forest Center for Integrative Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | | | | | | - Peter Brink
- Department of Physiology and Biophysics, SUNY Stony Brook, Stony Brook, NY 11794, USA
| | | | | |
Collapse
|
27
|
Caplan AI, Hariri R. Body Management: Mesenchymal Stem Cells Control the Internal Regenerator. Stem Cells Transl Med 2015; 4:695-701. [PMID: 26019227 DOI: 10.5966/sctm.2014-0291] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/25/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED SummaryIt has been assumed that adult tissues cannot regenerate themselves. With the current understanding that every adult tissue has its own intrinsic progenitor or stem cell, it is now clear that almost all tissues have regenerative potential partially related to their innate turnover dynamics. Moreover, it appears that a separate class of local cells originating as perivascular cells appears to provide regulatory oversight for localized tissue regeneration. The management of this regeneration oversight has a profound influence on the use of specific cells for cell therapies as a health care delivery tool set. The multipotent mesenchymal stem cell (MSC), now renamed the medicinal signaling cell, predominantly arises from pericytes released from broken and inflamed blood vessels and appears to function as both an immunomodulatory and a regeneration mediator. MSCs are being tested for their management capabilities to produce therapeutic outcomes in more than 480 clinical trials for a wide range of clinical conditions. Local MSCs function by managing the body's primary repair and regeneration activities. Supplemental MSCs can be provided from either endogenous or exogenous sources of either allogeneic or autologous origin. This MSC-based therapy has the potential to change how health care is delivered. These medicinal cells are capable of sensing their surroundings. Also, by using its complex signaling circuitry, these cells organize site-specific regenerative responses as if these therapeutic cells were well-programmed modern computers. Given these facts, it appears that we are entering a new age of cellular medicine. SIGNIFICANCE This report is a perspective from an active scientist and an active entrepreneur and commercial leader. It is neither a comprehensive review nor a narrowly focused treatise. The broad themes and the analogy to the working component of a computer and that of a cell are meant to draw several important scientific principles and health care themes together into the thesis that regenerative medicine is a constant throughout life and its management is the next frontier of health care. Mesenchymal stem cells are used as the central connection in the broad theme, not as multipotent progenitors but rather as an important control element in the natural local regeneration process.
Collapse
Affiliation(s)
- Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA;
| | - Robert Hariri
- Celgene Cellular Therapeutics, Warren, New Jersey, USA
| |
Collapse
|
28
|
Bouet G, Marchat D, Cruel M, Malaval L, Vico L. In VitroThree-Dimensional Bone Tissue Models: From Cells to Controlled and Dynamic Environment. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:133-56. [DOI: 10.1089/ten.teb.2013.0682] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Guenaelle Bouet
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| | - David Marchat
- Center for Biomedical and Healthcare Engineering, Ecole Nationale Supérieure des Mines, CIS-EMSE, CNRS:UMR 5307, Saint-Etienne, France
| | - Magali Cruel
- University of Lyon, LTDS, UMR CNRS 5513, Ecole Centrale de Lyon, Ecully, France
| | - Luc Malaval
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| | - Laurence Vico
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| |
Collapse
|
29
|
Alexander PG, Gottardi R, Lin H, Lozito TP, Tuan RS. Three-dimensional osteogenic and chondrogenic systems to model osteochondral physiology and degenerative joint diseases. Exp Biol Med (Maywood) 2014; 239:1080-95. [PMID: 24994814 DOI: 10.1177/1535370214539232] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tissue engineered constructs have the potential to function as in vitro pre-clinical models of normal tissue function and disease pathogenesis for drug screening and toxicity assessment. Effective high throughput assays demand minimal systems with clearly defined performance parameters. These systems must accurately model the structure and function of the human organs and their physiological response to different stimuli. Musculoskeletal tissues present unique challenges in this respect, as they are load-bearing, matrix-rich tissues whose functionality is intimately connected to the extracellular matrix and its organization. Of particular clinical importance is the osteochondral junction, the target tissue affected in degenerative joint diseases, such as osteoarthritis (OA), which consists of hyaline articular cartilage in close interaction with subchondral bone. In this review, we present an overview of currently available in vitro three-dimensional systems for bone and cartilage tissue engineering that mimic native physiology, and the utility and limitations of these systems. Specifically, we address the need to combine bone, cartilage and other tissues to form an interactive microphysiological system (MPS) to fully capture the biological complexity and mechanical functions of the osteochondral junction of the articular joint. The potential applications of three-dimensional MPSs for musculoskeletal biology and medicine are highlighted.
Collapse
Affiliation(s)
- Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Ri.MED Foundation, Palermo, I-90133 Italy
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Thomas P Lozito
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| |
Collapse
|
30
|
Lin HH, Hwang SM, Wu SJ, Hsu LF, Liao YH, Sheen YS, Chuang WH, Huang SY. The osteoblastogenesis potential of adipose mesenchymal stem cells in myeloma patients who had received intensive therapy. PLoS One 2014; 9:e94395. [PMID: 24722177 PMCID: PMC3983165 DOI: 10.1371/journal.pone.0094395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/14/2014] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is characterized by advanced osteolytic lesions resulting from the activation of osteoclasts (OCs) and inhibition of osteoblasts (OBs). OBs are derived from mesenchymal stem cells (MSCs) from the bone marrow (BM), however the pool and function of BMMSCs in MM patients (MM-BMMSCs) are reduced by myeloma cells (MCs) and cytokines secreted from MCs and related anti-MM treatment. Such reduction in MM-BMMSCs currently cannot be restored by any means. Recently, genetic aberrations of MM-BMMSCs have been noted, which further impaired their differentiation toward OBs. We hypothesize that the MSCs derived from adipose tissue (ADMSCs) can be used as alternative MSC sources to enhance the pool and function of OBs. Therefore, the purpose of this study was to compare the osteogenesis ability of paired ADMSCs and BMMSCs in MM patients who had completed intensive therapy. Fifteen MM patients who had received bortezomib-based induction and autologous transplantation were enrolled. At the third month after the transplant, the paired ADMSCs and BMMSCs were obtained and cultured. Compared with the BMMSCs, the ADMSCs exhibited a significantly higher expansion capacity (100% vs 13%, respectively; P = .001) and shorter doubling time (28 hours vs 115 hours, respectively; P = .019). After inducing osteogenic differentiation, although the ALP activity did not differ between the ADMSCs and BMMSCs (0.78 U/µg vs 0.74±0.14 U/µg, respectively; P = .834), the ADMSCs still exhibited higher calcium mineralization, which was determined using Alizarin red S (1029 nmole vs 341 nmole, respectively; P = .001) and von Kossa staining (2.6 E+05 µm2 vs 5 E+04 µm2, respectively; P = .042), than the BMMSCs did. Our results suggested that ADMSCs are a feasible MSC source for enhancing the pool and function of OBs in MM patients who have received intensive therapy.
Collapse
Affiliation(s)
- Hsiu-Hsia Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Shang-Ju Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Lee-Feng Hsu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Yi-Hua Liao
- Department of Dermatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Shuan Sheen
- Department of Dermatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Hui Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shang-Yi Huang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
31
|
Al-Nbaheen M, Vishnubalaji R, Ali D, Bouslimi A, Al-Jassir F, Megges M, Prigione A, Adjaye J, Kassem M, Aldahmash A. Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential. Stem Cell Rev Rep 2013; 9:32-43. [PMID: 22529014 PMCID: PMC3563956 DOI: 10.1007/s12015-012-9365-8] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human stromal (mesenchymal) stem cells (hMSCs) are multipotent stem cells with ability to differentiate into mesoderm-type cells e.g. osteoblasts and adipocytes and thus they are being introduced into clinical trials for tissue regeneration. Traditionally, hMSCs have been isolated from bone marrow, but the number of cells obtained is limited. Here, we compared the MSC-like cell populations, obtained from alternative sources for MSC: adipose tissue and skin, with the standard phenotype of human bone marrow MSC (BM-MSCs). MSC from human adipose tissue (human adipose stromal cells (hATSCs)) and human skin (human adult skin stromal cells, (hASSCs) and human new-born skin stromal cells (hNSSCs)) grew readily in culture and the growth rate was highest in hNSSCs and lowest in hATSCs. Compared with phenotype of hBM-MSC, all cell populations were CD34−, CD45−, CD14−, CD31−, HLA-DR−, CD13+, CD29+, CD44+, CD73+, CD90+,and CD105+. When exposed to in vitro differentiation, hATSCs, hASSCs and hNSSCs exhibited quantitative differences in their ability to differentiate into adipocytes and to osteoblastic cells. Using a microarray-based approach we have unveiled a common MSC molecular signature composed of 33 CD markers including known MSC markers and several novel markers e.g. CD165, CD276, and CD82. However, significant differences in the molecular phenotype between these different stromal cell populations were observed suggesting ontological and functional differences. In conclusion, MSC populations obtained from different tissues exhibit significant differences in their proliferation, differentiation and molecular phenotype, which should be taken into consideration when planning their use in clinical protocols.
Collapse
Affiliation(s)
- May Al-Nbaheen
- Stem Cell Unit, Department of Anatomy 28, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Kingdom of Saudi Arabia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Buschmann J, Gao S, Härter L, Hemmi S, Welti M, Werner CML, Calcagni M, Cinelli P, Wanner GA. Yield and proliferation rate of adipose-derived stromal cells as a function of age, body mass index and harvest site-increasing the yield by use of adherent and supernatant fractions? Cytotherapy 2013; 15:1098-105. [PMID: 23800730 DOI: 10.1016/j.jcyt.2013.04.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/19/2013] [Accepted: 04/23/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS Adipose-derived stem cells are easily accessed and have a relatively high density compared with other mesenchymal stromal cells. Isolation protocols of adipose-derived stem cells (ASC) rely on the cell's ability to adhere to tissue culture plastic overnight. It was evaluated whether the floating ASC fractions are also of interest for cell-based therapies. In addition, the impact of age, body mass index (BMI) and harvest site was assessed. METHODS The surface protein profile with the use of flow cytometry, the cell yield and the doubling time of passages 4, 5 and 6 of ASC from 30 donors were determined. Adherent and supernatant fractions were compared. The impact of age, BMI and harvest site on cell yield and doubling times was determined. RESULTS Both adherent and supernatant fractions showed high mean fluorescence intensities for CD13, CD29, CD44, CD73, CD90 and CD105 and comparatively low mean fluorescence intensities for CD11b, CD62L, intracellular adhesion molecule-1 and CD34. Doubling times of adherent and supernatant fractions did not differ significantly. Whereas the old age group had a significantly lower cell yield compared with the middle aged group, BMI and harvest site had no impact on cell yield. Finally, doubling times for passages 4, 5 and 6 were not influenced by the age and BMI of the donors, nor the tissue-harvesting site. CONCLUSIONS The floating ASC fraction is an equivalent second cell source just like the adherent ASC fraction. Donor age, BMI and harvest site do not influence cell yield and proliferation rate.
Collapse
Affiliation(s)
- Johanna Buschmann
- Division of Plastic and Hand Surgery, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mesenchymal stromal cells: misconceptions and evolving concepts. Cytotherapy 2013; 15:140-5. [DOI: 10.1016/j.jcyt.2012.11.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 11/01/2012] [Accepted: 11/06/2012] [Indexed: 12/17/2022]
|
34
|
Watt SM, Gullo F, van der Garde M, Markeson D, Camicia R, Khoo CP, Zwaginga JJ. The angiogenic properties of mesenchymal stem/stromal cells and their therapeutic potential. Br Med Bull 2013; 108:25-53. [PMID: 24152971 PMCID: PMC3842875 DOI: 10.1093/bmb/ldt031] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Blood vessel formation is fundamental to development, while its dysregulation can contribute to serious disease. Expectations are that hundreds of millions of individuals will benefit from therapeutic developments in vascular biology. MSCs are central to the three main vascular repair mechanisms. SOURCES OF DATA Key recent published literature and ClinicalTrials.gov. AREAS OF AGREEMENT MSCs are heterogeneous, containing multi-lineage stem and partly differentiated progenitor cells, and are easily expandable ex vivo. There is no single marker defining native MSCs in vivo. Their phenotype is strongly determined by their specific microenvironment. Bone marrow MSCs have skeletal stem cell properties. Having a perivascular/vascular location, they contribute to vascular formation and function and might be harnessed to regenerate a blood supply to injured tissues. AREAS OF CONTROVERSY These include MSC origin, phenotype and location in vivo and their ability to differentiate into functional cardiomyocytes and endothelial cells or act as vascular stem cells. In addition their efficacy, safety and potency in clinical trials in relation to cell source, dose, delivery route, passage and timing of administration, but probably even more on the local preconditioning and the mechanisms by which they exert their effects. GROWING POINTS Understanding the origin and the regenerative environment of MSCs, and manipulating their homing properties, proliferative ability and functionality through drug discovery and reprogramming strategies are important for their efficacy in vascular repair for regenerative medicine therapies and tissue engineering approaches. AREAS TIMELY FOR DEVELOPING RESEARCH Characterization of MSCs' in vivo origins and biological properties in relation to their localization within tissue niches, reprogramming strategies and newer imaging/bioengineering approaches.
Collapse
Affiliation(s)
- Suzanne M Watt
- Stem Cell Research Laboratory, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | | | | | | | | | | | |
Collapse
|
35
|
Spaas JH, Guest DJ, Van de Walle GR. Tendon Regeneration in Human and Equine Athletes. Sports Med 2012; 42:871-90. [DOI: 10.1007/bf03262300] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Abu Kasim NH, Govindasamy V, Gnanasegaran N, Musa S, Pradeep PJ, Srijaya TC, Aziz ZACA. Unique molecular signatures influencing the biological function and fate of post-natal stem cells isolated from different sources. J Tissue Eng Regen Med 2012; 9:E252-66. [PMID: 23229816 DOI: 10.1002/term.1663] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 09/04/2012] [Accepted: 10/31/2012] [Indexed: 12/21/2022]
Abstract
The discovery of mesenchymal stem cells (MSCs) from a myriad of tissues has triggered the initiative of establishing tailor-made stem cells for disease-specific therapy. Nevertheless, lack of understanding on the inherent differential propensities of these cells may restrict their clinical outcome. Therefore, a comprehensive study was done to compare the proliferation, differentiation, expression of cell surface markers and gene profiling of stem cells isolated from different sources, viz. bone marrow, Wharton's jelly, adipose tissue and dental pulp. We found that although all MSCs were phenotypically similar to each other, Wharton's jelly (WJ) MSCs and dental pulp stem cells (DPSCs) were highly proliferative as compared to bone marrow (BM) MSCs and adipose tissue (AD) MSCs. Moreover, indistinguishable cell surface characteristics and differentiation capacity were confirmed to be similar among all cell types. Based on gene expression profiling, we postulate that BM-MSCs constitutively expressed genes related to inflammation and immunodulation, whereas genes implicated in tissue development were highly expressed in AD-MSCs. Furthermore, the transcriptome profiling of WJ-MSCs and DPSCs revealed an inherent bias towards the neuro-ectoderm lineage. Based on our findings, we believe that there is no unique master mesenchymal stem cell that is appropriate to treat all target diseases. More precisely, MSCs from different sources exhibit distinct and unique gene expression signatures that make them competent to give rise to specific lineages rather than others. Therefore, stem cells should be subjected to rigorous characterization and utmost vigilance needs to be adopted in order to choose the best cellular source for a particular disease.
Collapse
Affiliation(s)
- Noor Hayaty Abu Kasim
- Department of Conservative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vijayendran Govindasamy
- Hygieia Innovation Sdn. Bhd, Lot 1G-2G, Lanai Complex No.2, Persiaran Seri Perdana, Percint 10, Federal Territory of Putrajaya, Malaysia.,Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Nareshwaran Gnanasegaran
- Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Sabri Musa
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Padmaja Jayaprasad Pradeep
- Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Zeti Adura Che Ab Aziz
- Department of Conservative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Phinney DG. Functional heterogeneity of mesenchymal stem cells: implications for cell therapy. J Cell Biochem 2012; 113:2806-12. [PMID: 22511358 DOI: 10.1002/jcb.24166] [Citation(s) in RCA: 301] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The term mesenchymal stem cell (MSCs) was adopted in the 1990s to describe a population of bone-marrow-derived cells that demonstrated the capacity for tri-lineage differentiation at a clonal level. Research conducted during the ensuing decades has demonstrated that MSCs fulfill many functions in addition to connective tissue progenitors including contributing to the HSC niche and regulating the function of immune effector cells of both the innate and adaptive immune system. Despite these advances, fundamental aspects of MSC biology remain indeterminate. For example, the embryonic origin of MSCs and their niche in vivo remains a highly debated topic. More importantly, the mechanisms that regulate self-renewal and lineage specification have also been largely unexplored. The later is significant in that MSC population's exhibit considerable donor-to-donor and intra-population heterogeneity but knowledge regarding how different functional attributes of MSCs are specified at the population level is unknown. This poses significant obstacles in research and in efforts to develop clinical manufacturing protocols that reproducibly generate functionally equivalent MSC populations. Herein, I discuss data demonstrating that MSC populations are intrinsically heterogeneous, elaborate on the molecular basis for this heterogeneity, and discuss how heterogeneity impacts clinical manufacturing and the therapeutic potency of MSCs.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, USA.
| |
Collapse
|
38
|
Abstract
With the ageing of the population and the major advances in targeted drug treatments, there is in medicine a shift in attention from survival towards quality of life. Therefore new challenges are emerging in modern health care. Preventive and personalized medicine have been identified as key steps in this context. New targeted biologicals for musculoskeletal diseases such as chronic arthritis have entered daily clinical practice, thereby not only controlling symptoms and signs, inflammation and destruction, but also maintaining function of the joints. The last aspect is essential for the independence of the individual and critical for the quality of life. Since the lifespan of prosthetic devices will always remain limited, new treatment approaches to repair skeletal structures need to be devised for the young and middle aged individuals with skeletal and joint damage caused by either congenital, traumatic, or inflammatory conditions. It is believed that regenerative medicine and more specifically tissue engineering may fill this void to some extent. Indeed, recent cellular therapeutics and combination products, now resorting under a new regulatory class of Advanced Medicinal Therapeutic Products, provide indications that progress is being made with clinically relevant outcomes in well-defined patient populations. For osteoarthritis, a joint disease leading to joint decompensation, novel tissue engineering therapies are being explored and, although most of the developments are still in early phase clinical studies, there are sufficient positive signals to pursue these novel therapeutic approaches in clinics. This article is part of a Special Issue entitled "Osteoarthritis".
Collapse
Affiliation(s)
- Frank P Luyten
- Division of Rheumatology, Department of Musculoskeletal Sciences, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, Belgium.
| | | |
Collapse
|
39
|
Giai Via A, Frizziero A, Oliva F. Biological properties of mesenchymal Stem Cells from different sources. Muscles Ligaments Tendons J 2012; 2:154-162. [PMID: 23738292 PMCID: PMC3666517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mesenchymal stem cells (MSCs) are adult, nonhematopoietic, stem cells that were initially isolated from bone marrow. Now they can be isolated from almost every tissue of the body. They have the ability to self-renew and differentiate into multiple cell lineage, including bone, chondrocytes, adipocytes, tenocytes and cardiomyocytes, and it makes them an attractive cell source for a new generation of cell-based regenerative therapies. In this review we try to summarize data on sources and the biological properties of MSCs.
Collapse
Affiliation(s)
- Alessio Giai Via
- Department of Orthopaedics and Traumatology, University of Rome “Tor Vergata” School of Medicine, Rome, Italy
| | - Antonio Frizziero
- Department of Orthopaedic Rehabilitation, University of Padova, Italy
| | - Francesco Oliva
- Department of Orthopaedics and Traumatology, University of Rome “Tor Vergata” School of Medicine, Rome, Italy
| |
Collapse
|
40
|
Abstract
INTRODUCTION The biological process of fracture healing is complex with influences that are both patient-dependent and related to the trauma experienced and stability of the fracture. Fracture healing complications negatively affect the patient's quality of life, even more when fractures occur in the elderly osteoporotic patients. AREAS COVERED In the polytherapy for bone regeneration, a high success rate was obtained with the use of growth factors, osteogenic cells, and osteoconductive factors. There have been high expectations that treatment with drugs active on bone remodeling would be efficient for acceleration of fracture healing. A literature search was undertaken using wording like "drug or pharmacology of fracture healing." This report will review the systemic pharmacological agents for which clinical trials documenting their efficacy on bone healing have been carried out or are underway. EXPERT OPINION At present the use of systemic pharmacological agents to enhance fracture healing in the clinical setting is still controversial. However, future clinical trials will offer the possibility to obtain data that will make possible the registration of a drug as a "healer."
Collapse
Affiliation(s)
- Maria Luisa Brandi
- University of Florence, Department of Surgery and Translational Medicine, Mineral and Bone Metabolic Diseases Unit, Largo Palagi, 1, 50100 Florence, Italy.
| |
Collapse
|
41
|
Lubelski D, Abdullah KG, Benzel EC, Mroz TE. The Utility of Allograft Mesenchymal Stem Cells for Spine Fusion: A Literature Review. Global Spine J 2012; 2:109-14. [PMID: 27054055 PMCID: PMC4813091 DOI: 10.1055/s-0032-1307263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
More than 50% of patients complain of postoperative donor site morbidity following iliac crest bone graft harvest, and recent discoveries have identified adverse outcomes following bone morphogenetic protein use in spine fusion. This has led the spine community to turn toward alternative methods to promote fusion following spine surgery. The present article reviews numerous studies that have shown the osteogenic potential of mesenchymal stem cells (MSCs). MSCs have been used with both in vitro and in vivo models and have involved animal studies ranging from rats to macaque monkeys to successfully induce bone regeneration in lesions of the tibia and spine. There is no fear of graft rejection, as there may be with other allograft materials, because neither undifferentiated nor differentiated MSCs elicit lymphocyte response when transplanted; they tend to alter the cytokine profile to an anti-inflammatory state. Early clinical trials are underway with various commercially available MSC formulations. Although there is much enthusiasm, it is integral that the spine surgery community carefully evaluate the use of MSCs in spine fusion through well-designed and executed studies to determine the efficacy and safety profiles in spine surgery patients.
Collapse
Affiliation(s)
- Daniel Lubelski
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Kalil G. Abdullah
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Edward C. Benzel
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Thomas E. Mroz
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| |
Collapse
|
42
|
Karantalis V, Balkan W, Schulman IH, Hatzistergos KE, Hare JM. Cell-based therapy for prevention and reversal of myocardial remodeling. Am J Physiol Heart Circ Physiol 2012; 303:H256-70. [PMID: 22636682 DOI: 10.1152/ajpheart.00221.2012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although pharmacological and interventional advances have reduced the morbidity and mortality of ischemic heart disease, there is an ongoing need for novel therapeutic strategies that prevent or reverse progressive ventricular remodeling following myocardial infarction, the process that forms the substrate for ventricular failure. The development of cell-based therapy as a strategy to repair or regenerate injured tissue offers extraordinary promise for a powerful anti-remodeling therapy. In this regard, the field of cell therapy has made major advancements in the past decade. Accumulating data from preclinical studies have provided novel insights into stem cell engraftment, differentiation, and interactions with host cellular elements, as well as the effectiveness of various methods of cell delivery and accuracy of diverse imaging modalities to assess therapeutic efficacy. These findings have in turn guided rationally designed translational clinical investigations. Collectively, there is a growing understanding of the parameters that underlie successful cell-based approaches for improving heart structure and function in ischemic and other cardiomyopathies.
Collapse
Affiliation(s)
- Vasileios Karantalis
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, USA
| | | | | | | | | |
Collapse
|
43
|
Williams EL, Edwards CJ, Cooper C, Oreffo ROC. The osteoarthritic niche and modulation of skeletal stem cell function for regenerative medicine. J Tissue Eng Regen Med 2012; 7:589-608. [PMID: 22489025 DOI: 10.1002/term.1455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 10/18/2011] [Accepted: 11/24/2011] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is the most common cause of arthritis worldwide and represents a significant healthcare burden, particularly in the context of an ageing population. Traditionally, painkillers, injections and physiotherapy have been the mainstay of treatment, with patients being referred for joint replacement surgery (arthroplasty) when these options fail. Whilst effective in reducing pain and improving joint function, these approaches are not without potential complications. With the development of tissue-engineering techniques over recent years there has been considerable interest in applying these strategies to provide new, innovative, alternative effective means of treating OA. This review explores the unique microenvironment present within an osteoarthritic joint, highlighting the features that comprise the osteoarthritic niche and could be modulated in the development of novel treatments for OA. Existing tissue-engineering strategies for repairing bone and cartilage defects are discussed, with particular reference to how these might be modified, both to improve existing treatments, such as impaction bone grafting, as well as in the development of future treatments for OA.
Collapse
Affiliation(s)
- E L Williams
- Bone and Joint Research Group, Human Development and Health, University of Southampton Medical School, UK.
| | | | | | | |
Collapse
|
44
|
Smith JO, Aarvold A, Tayton ER, Dunlop DG, Oreffo ROC. Skeletal tissue regeneration: current approaches, challenges, and novel reconstructive strategies for an aging population. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:307-20. [PMID: 21615329 DOI: 10.1089/ten.teb.2011.0143] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Loss of skeletal tissue as a consequence of trauma, injury, or disease is a significant cause of morbidity with often wide-ranging socioeconomic impacts. Current approaches to replace or restore significant quantities of lost bone come with substantial limitations and inherent disadvantages that may in themselves cause further disability. In addition, the spontaneous repair capacity of articular cartilage is limited; thus, investigation into new cartilage replacement and regeneration techniques are warranted. Along with the challenges of an increasingly aging demographic, changing clinical scenarios and rising functional expectations provide the imperative for new, more reliable skeletal regeneration strategies. The science of tissue engineering has expanded dramatically in recent years, notably in orthopedic applications, and it is clear that new approaches for de novo skeletal tissue formation offer exciting opportunities to improve the quality of life for many, particularly in the face of increasing patient expectations. However, significant scientific, financial, industrial, and regulatory challenges should be overcome before the successful development of an emergent tissue engineering strategy can be realized. We outline current practice for replacement of lost skeletal tissue and the innovative approaches in tissue regeneration that have so far been translated to clinical use, along with a discussion of the significant hurdles that are presented in the process of translating research strategies to the clinic.
Collapse
Affiliation(s)
- James Oliver Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, Southampton, United Kingdom
| | | | | | | | | |
Collapse
|
45
|
Abstract
Improving bone repair remains an important and challenging issue. Therapeutic approaches to amplify osteogenic cell pool or function include cell and gene therapies. We identified genes that promote human mesenchymal cell differentiation and bone formation. Targeting these or other genes may improve the efficacy of cell therapy for bone repair.
Collapse
Affiliation(s)
- P J Marie
- Laboratory of Osteoblast Biology and Pathology, Inserm UMR606 and University Paris Diderot, Hopital Lariboisiere, 2 rue Ambroise Pare, 75475 Paris cedex 10, France.
| |
Collapse
|
46
|
Sévère N, Miraoui H, Marie PJ. The Casitas B lineage lymphoma (Cbl) mutant G306E enhances osteogenic differentiation in human mesenchymal stromal cells in part by decreased Cbl-mediated platelet-derived growth factor receptor alpha and fibroblast growth factor receptor 2 ubiquitination. J Biol Chem 2011; 286:24443-50. [PMID: 21596750 DOI: 10.1074/jbc.m110.197525] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human bone marrow-derived mesenchymal stromal cells (hMSCs) have the capacity to differentiate into several cell types including osteoblasts and are therefore an important cell source for bone tissue regeneration. A crucial issue is to identify mechanisms that trigger hMSC osteoblast differentiation to promote osteogenic potential. Casitas B lineage lymphoma (Cbl) is an E3 ubiquitin ligase that ubiquitinates and targets several molecules for degradation. We hypothesized that attenuation of Cbl-mediated degradation of receptor tyrosine kinases (RTKs) may promote osteogenic differentiation in hMSCs. We show here that specific inhibition of Cbl interaction with RTKs using a Cbl mutant (G306E) promotes expression of osteoblast markers (Runx2, alkaline phosphatase, type 1 collagen, osteocalcin) and increases osteogenic differentiation in clonal bone marrow-derived hMSCs and primary hMSCs. Analysis of molecular mechanisms revealed that the Cbl mutant increased PDGF receptor α and FGF receptor 2 but not EGF receptor expression in hMSCs, resulting in increased ERK1/2 and PI3K signaling. Pharmacological inhibition of FGFR or PDGFR abrogated in vitro osteogenesis induced by the Cbl mutant. The data reveal that specific inhibition of Cbl interaction with RTKs promotes the osteogenic differentiation program in hMSCs in part by decreased Cbl-mediated PDGFRα and FGFR2 ubiquitination, providing a novel mechanistic approach targeting Cbl to promote the osteogenic capacity of hMSCs.
Collapse
Affiliation(s)
- Nicolas Sévère
- Laboratory of Osteoblast Biology and Pathology, INSERM, U606, Paris F-75475, France
| | | | | |
Collapse
|
47
|
Abstract
This brief overview summarises the main characteristics of bone marrow mesenchymal stem cells and of adipose-derived stem cells: methods of obtention, phenotype, differentiation potential, hematopoiesis-supportive (stromal) capacity, and immunosuppressive properties. Two points are discussed in detail: 1) criteria for stemness: multipotency, self-renewal, plasticity, and 2) the repair mechanisms implicated in the different indications of cell therapy using these cells: reconstitution of the tissue functional compartment by repopulation consequent to proliferation and differentiation or reprogrammation, stromal effects by secretion of angiogenic, anti-apoptotic, anti-fibrogenic factors, molecules involved in the regulation of inflammation, etc.
Collapse
Affiliation(s)
- Pierre Charbord
- Inserm U972, bâtiment Gregory Pincus, hôpital de Bicêtre, 80, avenue du Général Leclerc, 94276 Le Kremlin Bicêtre Cedex, France.
| | | |
Collapse
|
48
|
Charbord P. Bone marrow mesenchymal stem cells: historical overview and concepts. Hum Gene Ther 2010; 21:1045-56. [PMID: 20565251 DOI: 10.1089/hum.2010.115] [Citation(s) in RCA: 285] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This review describes the historical emergence of the concept of bone marrow mesenchymal stem cells (MSCs), summarizing data on Wolf and Trentin's hematopoietic inductive microenvironment; Dexter's hematopoiesis-supportive stromal cells; Friedenstein's osteogenic cells; and Pittenger's trilineal osteoblastic, chondrocytic, and adipocytic precursors; to finally introduce the specific bone marrow mesenchymal stem cells with differentiation potential to four lineages (mesenchymal and vascular smooth muscle lineages), and stromal and immunomodulatory capacities. Two points are the object of detailed discussion. The first point envisions the stem cell attributes (multipotentiality, self-renewal, tissue regeneration, population heterogeneity, plasticity, and lineage priming) compared with that of the paradigmatic hematopoietic stem cell. In the second point, we discuss the possible existence of bone marrow cells with greater differentiation potential, eventually pluripotential cells. The latter point raises the issues of cell fusion, reprogramming, or selection under nonstandardized conditions of rare populations of neuroectodermal origin, or of cells that had undergone mesenchymal-to-epithelial transition. In the last section, we review data on MSC senescence and possible malignant transformation secondary to extensive culture, gene transfer of telomerase, or mutations such as leading to Ewing's sarcoma. The set of data leads to the conclusion that bone marrow MSCs constitute a specific adult tissue stem cell population. The multiple characteristics of this stem cell type account for the versatility of the mechanisms of injured tissue repair. Although MSC administration may be extremely useful in a number of clinical applications, their transplantation is not without risks that must not be overlooked when developing cell therapy protocols.
Collapse
Affiliation(s)
- Pierre Charbord
- Institut National de la Recherche et Santé Médicale U, Université Paris XI, Kremlin Bicêtre, France.
| |
Collapse
|
49
|
Hocking AM, Gibran NS. Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res 2010; 316:2213-9. [PMID: 20471978 DOI: 10.1016/j.yexcr.2010.05.009] [Citation(s) in RCA: 269] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 05/07/2010] [Accepted: 05/08/2010] [Indexed: 12/13/2022]
Abstract
Cutaneous wounds persist as a health care crisis in spite of increased understanding of the cellular and molecular responses to injury. Contributing significantly to this crisis is the lack of reliable therapies for treatment of wounds that are slow to heal including chronic wounds and deep dermal wounds that develop hypertrophic scars. This article will review the growing evidence demonstrating the promise of multipotent mesenchymal stem/stromal (MSCs) for the treatment of impaired wound healing. MSCs are often referred to as mesenchymal stem cells despite concerns that these cells are not truly stem cells given the lack of evidence demonstrating self-renewal in vivo. Regardless, abundant evidence demonstrates the therapeutic potential of MSCs for repair and regeneration of damaged tissue due to injury or disease. To date, MSC treatment of acute and chronic wounds results in accelerated wound closure with increased epithelialization, granulation tissue formation and angiogenesis. Although there is evidence for MSC differentiation in the wound, most of the therapeutic effects are likely due to MSCs releasing soluble factors that regulate local cellular responses to cutaneous injury. Important challenges need to be overcome before MSCs can be used effectively to treat wounds that are slow to heal.
Collapse
Affiliation(s)
- Anne M Hocking
- University of Washington, Campus Box 359796, 300 9th Avenue, Seattle, WA 98104, USA.
| | | |
Collapse
|
50
|
Backly RME, Cancedda R. Bone marrow stem cells in clinical application: harnessing paracrine roles and niche mechanisms. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 123:265-92. [PMID: 20803145 DOI: 10.1007/10_2010_78] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The being of any individual throughout life is a dynamic process relying on the capacity to retain processes of self-renewal and differentiation, both of which are hallmarks of stem cells. Although limited in the adult human organism, regeneration and repair do take place in virtue of the presence of adult stem cells. In the bone marrow, two major populations of stem cells govern the dynamic equilibrium of both hemopoiesis and skeletal homeostasis; the hematopoietic and the mesenchymal stem cells. Recent cell based clinical trials utilizing bone marrow-derived stem cells as therapeutic agents have revealed promising results, while others have failed to display as such. It is therefore imperative to strive to understand the mechanisms by which these cells function in vivo, how their properties can be maintained ex-vivo, and to explore further their recently highlighted immunomodulatory and trophic effects.
Collapse
Affiliation(s)
- Rania M El Backly
- Istituto Nazionale per la Ricerca sul Cancro, and Dipartimento di Oncologia, Biologia e Genetica dell'Universita' di Genova, Genova, Italy
| | | |
Collapse
|