51
|
Molecular Mechanisms Responsible for Anti-inflammatory and Immunosuppressive Effects of Mesenchymal Stem Cell-Derived Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:187-206. [PMID: 31175638 DOI: 10.1007/5584_2018_306] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are self-renewable cells capable for multilineage differentiation and immunomodulation. MSCs are able to differentiate into all cell types of mesodermal origin and, due to their plasticity, may generate cells of neuroectodermal or endodermal origin in vitro. In addition to the enormous differentiation potential, MSCs efficiently modulate innate and adaptive immune response and, accordingly, were used in large number of experimental and clinical trials as new therapeutic agents in regenerative medicine. Although MSC-based therapy was efficient in the treatment of many inflammatory and degenerative diseases, unwanted differentiation of engrafted MSCs represents important safety concern. MSC-based beneficial effects are mostly relied on the effects of MSC-derived immunomodulatory, pro-angiogenic, and trophic factors which attenuate detrimental immune response and inflammation, reduce ischemic injuries, and promote tissue repair and regeneration. Accordingly, MSC-conditioned medium (MSC-CM), which contains MSC-derived factors, has the potential to serve as a cell-free, safe therapeutic agent for the treatment of inflammatory diseases. Herein, we summarized current knowledge regarding identification, isolation, ontogeny, and functional characteristics of MSCs and described molecular mechanisms responsible for MSC-CM-mediated anti-inflammatory and immunosuppressive effects in the therapy of inflammatory lung, liver, and kidney diseases and ischemic brain injury.
Collapse
|
52
|
Casiraghi F, Perico N, Remuzzi G. Mesenchymal stromal cells for tolerance induction in organ transplantation. Hum Immunol 2017; 79:304-313. [PMID: 29288697 DOI: 10.1016/j.humimm.2017.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/06/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022]
Abstract
The primary challenge in organ transplantation continues to be the need to suppress the host immune system long-term to ensure prolonged allograft survival. Long-term non-specific immunosuppression can, however, result in life-threatening complications. Thus, efforts have been pursued to explore novel strategies that would allow minimization of maintenance immunosuppression, eventually leading to transplant tolerance. In this scenario, bone marrow-derived mesenchymal stromal cells (MSC), given their unique immunomodulatory properties to skew the balance between regulatory and memory T cells, have emerged as potential candidates for cell-based therapy to promote immune tolerance. Here, we review our initial clinical experience with bone marrow-derived MSC in living-donor kidney transplant recipients and provide an overview of the available results of other clinical programs with MSC in kidney and liver transplantation, highlighting hurdles and success of this innovative cell-based therapy.
Collapse
Affiliation(s)
| | - Norberto Perico
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy; Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale (ASST), Papa Giovanni XXIII, Bergamo, Italy; L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
53
|
Furuhata Y, Nihongaki Y, Sato M, Yoshimoto K. Control of Adipogenic Differentiation in Mesenchymal Stem Cells via Endogenous Gene Activation Using CRISPR-Cas9. ACS Synth Biol 2017; 6:2191-2197. [PMID: 29077398 DOI: 10.1021/acssynbio.7b00246] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cells (MSCs) are of interest in regenerative medicine owing to their multilineage differentiation and self-renewal properties. Understanding the in vivo differentiation process is necessary for clinical applications including cell therapy and transplantation. This remains challenging owing to the lack of induction methods that imitate the natural programming process. Endogenous gene regulation of tissue-specific transcription factors is therefore desirable. In the present study, we demonstrated endogenous activation of adipogenic genes through the dCas9-based transcription system and achieved efficient induction of different types of adipocyte-like cells from MSCs. Interestingly, the MSCs converted via single-gene activation exhibited morphological and molecular properties of white adipocytes, while beige adipocyte-like cells were induced via multiplex gene activation of three specific transcription factors. These results reveal that the fate of MSCs can be effectively manipulated by direct activation of specific endogenous gene expression using a dCas9-based activator with reduced exogenous additives.
Collapse
Affiliation(s)
- Yuichi Furuhata
- Department
of Computational Biology and Medical Sciences, Graduate School of
Frontier Sciences, The University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Japan
| | - Yuta Nihongaki
- Department
of General Systems Studies, Graduate School of Arts and Sciences, The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo 153-8902, Japan
| | - Moritoshi Sato
- Department
of General Systems Studies, Graduate School of Arts and Sciences, The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo 153-8902, Japan
| | - Keitaro Yoshimoto
- Department
of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo 153-8902, Japan
- JST,
PRESTO, The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
54
|
Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl Med 2017; 6:2173-2185. [PMID: 29076267 PMCID: PMC5702523 DOI: 10.1002/sctm.17-0129] [Citation(s) in RCA: 505] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) hold great potential for regenerative medicine because of their ability for self-renewal and differentiation into tissue-specific cells such as osteoblasts, chondrocytes, and adipocytes. MSCs orchestrate tissue development, maintenance and repair, and are useful for musculoskeletal regenerative therapies to treat age-related orthopedic degenerative diseases and other clinical conditions. Importantly, MSCs produce secretory factors that play critical roles in tissue repair that support both engraftment and trophic functions (autocrine and paracrine). The development of uniform protocols for both preparation and characterization of MSCs, including standardized functional assays for evaluation of their biological potential, are critical factors contributing to their clinical utility. Quality control and release criteria for MSCs should include cell surface markers, differentiation potential, and other essential cell parameters. For example, cell surface marker profiles (surfactome), bone-forming capacities in ectopic and orthotopic models, as well as cell size and granularity, telomere length, senescence status, trophic factor secretion (secretome), and immunomodulation, should be thoroughly assessed to predict MSC utility for regenerative medicine. We propose that these and other functionalities of MSCs should be characterized prior to use in clinical applications as part of comprehensive and uniform guidelines and release criteria for their clinical-grade production to achieve predictably favorable treatment outcomes for stem cell therapy. Stem Cells Translational Medicine 2017;6:2173-2185.
Collapse
Affiliation(s)
- Rebekah M. Samsonraj
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Department of Orthopaedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Michael Raghunath
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Center for Cell Biology and Tissue Engineering, Competence Center for Tissue Engineering and Substance Testing (TEDD)Institute for Chemistry and Biotechnology, ZHAW School of Life Sciences and Facility Management, Zurich University of Applied SciencesSwitzerland
| | - Victor Nurcombe
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
| | - James H. Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | | | - Simon M. Cool
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| |
Collapse
|
55
|
Perico N, Casiraghi F, Remuzzi G. Clinical Translation of Mesenchymal Stromal Cell Therapies in Nephrology. J Am Soc Nephrol 2017; 29:362-375. [PMID: 29191959 DOI: 10.1681/asn.2017070781] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells have emerged as potential candidates for cell-based therapies to modulate the immune response in organ transplantation and repair tissues after acute or chronic injury. Preclinical studies have shown convincingly in rodent models that mesenchymal stromal cells can prolong solid organ graft survival and that they can induce immune tolerance, accelerate recovery from AKI, and promote functional improvement in chronic nephropathies. Multiple complex properties of the cells, including immunomodulatory, anti-inflammatory, and proregenerative effects, seem to contribute. The promising preclinical studies have encouraged investigators to explore the safety, tolerability, and efficacy of mesenchymal stromal cell-based therapy in pilot clinical trials, including those for bone marrow and solid organ transplantation, autoimmune diseases, and tissue and organ repair. Here, we review the available data on culture-expanded mesenchymal stromal cells tested in renal transplantation, AKI, and CKD. We also briefly discuss the relevant issues that must be addressed to ensure rigorous assessment of the safety and efficacy of mesenchymal stromal cell therapies to allow the translation of this research into the practice of clinical nephrology.
Collapse
Affiliation(s)
- Norberto Perico
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy;
| | - Federica Casiraghi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Giuseppe Remuzzi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy.,Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy; and.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
56
|
Kizilay Mancini O, Lora M, Cuillerier A, Shum-Tim D, Hamdy R, Burelle Y, Servant MJ, Stochaj U, Colmegna I. Mitochondrial Oxidative Stress Reduces the Immunopotency of Mesenchymal Stromal Cells in Adults With Coronary Artery Disease. Circ Res 2017; 122:255-266. [PMID: 29113965 DOI: 10.1161/circresaha.117.311400] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 12/25/2022]
Abstract
RATIONALE Mesenchymal stromal cells (MSCs) are promising therapeutic strategies for coronary artery disease; however, donor-related variability in cell quality is a main cause of discrepancies in preclinical studies. In vitro, MSCs from individuals with coronary artery disease have reduced ability to suppress activated T-cells. The mechanisms underlying the altered immunomodulatory capacity of MSCs in the context of atherosclerosis remain elusive. OBJECTIVE The aim of this study was to assess the role of mitochondrial dysfunction in the impaired immunomodulatory properties of MSCs from patients with atherosclerosis. METHODS AND RESULTS Adipose tissue-derived MSCs were isolated from atherosclerotic (n=38) and nonatherosclerotic (n=42) donors. MSCs:CD4+T-cell suppression was assessed in allogeneic coculture systems. Compared with nonatherosclerotic-MSCs, atherosclerotic-MSCs displayed higher levels of both intracellular (P=0.006) and mitochondrial (P=0.03) reactive oxygen species reflecting altered mitochondrial function. The increased mitochondrial reactive oxygen species levels of atherosclerotic-MSCs promoted a phenotypic switch characterized by enhanced glycolysis and an altered cytokine secretion (interleukin-6 P<0.0001, interleukin-8/C-X-C motif chemokine ligand 8 P=0.04, and monocyte chemoattractant protein-1/chemokine ligand 2 P=0.01). Furthermore, treatment of atherosclerotic-MSCs with the reactive oxygen species scavenger N-acetyl-l-cysteine reduced the levels of interleukin-6, interleukin-8/C-X-C motif chemokine ligand 8, and monocyte chemoattractant protein-1/chemokine ligand 2 in the MSC secretome and improved MSCs immunosuppressive capacity (P=0.03). CONCLUSIONS An impaired mitochondrial function of atherosclerotic-MSCs underlies their altered secretome and reduced immunopotency. Interventions aimed at restoring the mitochondrial function of atherosclerotic-MSCs improve their in vitro immunosuppressive ability and may translate into enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Ozge Kizilay Mancini
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Maximilien Lora
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Alexanne Cuillerier
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Dominique Shum-Tim
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Reggie Hamdy
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Yan Burelle
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Marc J Servant
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Ursula Stochaj
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada
| | - Inés Colmegna
- From the Department of Anatomy and Cell Biology (O.K.M.), Department of Physiology (U.S.), Divisions of Cardiac Surgery and Surgical Research, Department of Surgery (D.S.T.), Division of Rheumatology, Department of Medicine (I.C., M.L.) McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children (R.H.); Department of Cellular and Molecular Medicine, Faculty of Medicine (A.C., Y.B.), University of Ottawa, Ontario, Canada; and Faculty of Pharmacy (M.J.S.), University of Montreal, Quebec, Canada.
| |
Collapse
|
57
|
Cryopreserved or Fresh Mesenchymal Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Potential of MSC Therapy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:77-98. [PMID: 27837556 DOI: 10.1007/978-3-319-45457-3_7] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) harbor great therapeutic potential for numerous diseases. From early clinical trials, success and failure analysis, bench-to-bedside and back-to-bench approaches, there has been a great gain in knowledge, still leaving a number of questions to be answered regarding optimal manufacturing and quality of MSCs for clinical application. For treatment of many acute indications, cryobanking may remain a prerequisite, but great uncertainty exists considering the therapeutic value of freshly thawed (thawed) and continuously cultured (fresh) MSCs. The field has seen an explosion of new literature lately, outlining the relevance of the topic. MSCs appear to have compromised immunomodulatory activity directly after thawing for clinical application. This may provide a possible explanation for failure of early clinical trials. It is not clear if and how quickly MSCs recover their full therapeutic activity, and if the "cryo stun effect" is relevant for clinical success. Here, we will share our latest insights into the relevance of these observations for clinical practice that will be discussed in the context of the published literature. We argue that the differences of fresh and thawed MSCs are limited but significant. A key issue in evaluating potency differences is the time point of analysis after thawing. To date, prospective double-blinded randomized clinical studies to evaluate potency of both products are lacking, although recent progress was made with preclinical assessment. We suggest refocusing therapeutic MSC development on potency and safety assays with close resemblance of the clinical reality.
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW Mesenchymal stromal cells (MSCs) are adult stromal cells with therapeutic potential in allogeneic islet transplantation for type 1 diabetes patients. The process of islet isolation alone has been shown to negatively impact islet survival and function in vivo. In addition, insults mediated by the instant blood-mediated inflammatory reaction, hypoxia, ischemia and immune response significantly impact the islet allograft post transplantation. MSCs are known to exert cytoprotective and immune modulatory properties and thus are an attractive therapeutic in this context. Herein, the recent progress in the field of MSC therapy in islet transplantation is discussed. RECENT FINDINGS MSC can promote islet survival and function in vivo. Importantly, studies have shown that human MSC donors have differential abilities in promoting islet regeneration/survival. Recently, several biomarkers associated with MSC islet regenerative capacity have been identified. Expressions of Annexin A1, Elastin microfibril interface 1 and integrin-linked protein kinase are upregulated in MSC displaying protective effects on islet survival and function in vivo. SUMMARY The discovery of biomarkers associated with MSC therapeutic efficacy represents an important step forward for the utilization of MSC therapy in islet transplantation; however, much remains to be elucidated about the mechanisms utilized by MSC in protection against transplanted islet loss, autoimmune-mediated and alloimmune-mediated rejection.
Collapse
|
59
|
Mesenchymal stem cell therapy to promote corneal allograft survival: current status and pathway to clinical translation. Curr Opin Organ Transplant 2017; 21:559-567. [PMID: 27801687 DOI: 10.1097/mot.0000000000000360] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This article reviews the literature on the therapeutic potential of mesenchymal stem cells (MSCs) to prolong corneal allograft survival. RECENT FINDINGS To date, only small numbers studies have investigated the MSC ability to modulate corneal allograft survival. Most reports have shown positive results, which is encouraging, however as different MSC-application strategies (time point of injection, cell number/number of injections, route of injection, MSC source, MSC licensing) have been employed in various animal models it is difficult to compare and validate the results. The MSC ability to promote graft survival has been attributed to their modulation of the recipient immune system, altering the Th1/Th2 balance, expanding Foxp3 regulatory T cells, polarizing macrophages and inhibiting intra-graft infiltration of antigen presenting cells. More in depth analysis is required to elucidate the mechanism of MSC-immunomodulation in vivo. SUMMARY MSCs have shown the potential to modulate corneal allograft rejection in various models using MSCs from different species. In particular for high-risk patients with poor prognosis MSC therapy might be a promising approach to promote corneal allograft survival. First-in-man clinical trials with MSC will hopefully shed new light on MSC-mediated immunomodulation in vivo and contribute to the restoration of vision in patients receiving corneal allografts.
Collapse
|
60
|
Song JY, Kang HJ, Hong JS, Kim CJ, Shim JY, Lee CW, Choi J. Umbilical cord-derived mesenchymal stem cell extracts reduce colitis in mice by re-polarizing intestinal macrophages. Sci Rep 2017; 7:9412. [PMID: 28842625 PMCID: PMC5573412 DOI: 10.1038/s41598-017-09827-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/31/2017] [Indexed: 12/16/2022] Open
Abstract
Human umbilical cord mesenchymal stem cells (hUC-MSCs), originating in Wharton’s jelly, are multipotent stem cells that home to damaged tissues and can modulate the immune system. We examined whether administering extracts of MSCs (MSC-Ex) instead of MSCs could augment the beneficial effects of MSC therapy by overcoming the low homing efficiency of MSCs systemically administered in inflammatory bowel diseases (IBD). Dextran sodium sulfate-induced colitis model was established in C57BL/6 mice, and MSC-Ex was administered intraperitoneally. MSC-Ex reduced colitis, disease activity index (DAI), and histological colitis scores, and increased the body weight. Treatment with MSC-Ex completely blocked the induction of inflammatory cytokines, which were strongly detected in mice with colitis. MSC-Ex shifted the macrophage functional phenotype from M1 to M2 by decreasing the levels of MCP1, CXCL9, and iNOS, but increasing the levels of IL-10, LIGHT, CCL1, and Arg-1. MSC-Ex recovered the destruction of the epithelial barrier in the differentiated Caco-2 cells in vitro. Treatment with MSC-Ex was more potent than that with MSC in reducing DAI, the histological score, and nitrite levels. These data strongly support that MSC-Ex treatment can be a potent approach to overcome severe refractory IBD.
Collapse
Affiliation(s)
- Ji-Young Song
- Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Hyo Jeong Kang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Department of Physiology, Asan-Minnesota Institute for Innovating Transplantation, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Joon Seok Hong
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Gyeonggi-do, Korea
| | - Chong Jai Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jae-Yoon Shim
- Department Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Christopher W Lee
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, California, USA
| | - Jene Choi
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
61
|
Nagaishi K, Mizue Y, Chikenji T, Otani M, Nakano M, Saijo Y, Tsuchida H, Ishioka S, Nishikawa A, Saito T, Fujimiya M. Umbilical cord extracts improve diabetic abnormalities in bone marrow-derived mesenchymal stem cells and increase their therapeutic effects on diabetic nephropathy. Sci Rep 2017; 7:8484. [PMID: 28814814 PMCID: PMC5559488 DOI: 10.1038/s41598-017-08921-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 07/20/2017] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSC) has been applied as the most valuable source of autologous cell transplantation for various diseases including diabetic complications. However, hyperglycemia may cause abnormalities in intrinsic BM-MSC which might lose sufficient therapeutic effects in diabetic patients. We demonstrated the functional abnormalities in BM-MSC derived from both type 1 and type 2 diabetes models in vitro, which resulted in loss of therapeutic effects in vivo in diabetic nephropathy (DN). Then, we developed a novel method to improve abnormalities in BM-MSC using human umbilical cord extracts, namely Wharton’s jelly extract supernatant (WJs). WJs is a cocktail of growth factors, extracellular matrixes and exosomes, which ameliorates proliferative capacity, motility, mitochondrial degeneration, endoplasmic reticular functions and exosome secretions in both type 1 and type 2 diabetes-derived BM-MSC (DM-MSC). Exosomes contained in WJs were a key factor for this activation, which exerted similar effects to complete WJs. DM-MSC activated by WJs ameliorated renal injury in both type 1 and type 2 DN. In this study, we developed a novel activating method using WJs to significantly increase the therapeutic effect of BM-MSC, which may allow effective autologous cell transplantation.
Collapse
Affiliation(s)
- Kanna Nagaishi
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan. .,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan.
| | - Yuka Mizue
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Takako Chikenji
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Miho Otani
- Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| | - Masako Nakano
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Yusaku Saijo
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Hikaru Tsuchida
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan
| | - Shinichi Ishioka
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Akira Nishikawa
- Department of Gynecology and Obstetrics, NTT Sapporo Hospital, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Mineko Fujimiya
- Second Department of Anatomy, Sapporo Medical University, Sapporo, Japan.,Department of Diabetic Cellular Therapeutics, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
62
|
Kouroupis D, Wang XN, El-Sherbiny Y, McGonagle D, Jones E. The Safety of Non-Expanded Multipotential Stromal Cell Therapies. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-3-319-59165-0_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
63
|
Zheng G, Qiu G, Ge M, He J, Huang L, Chen P, Wang W, Xu Q, Hu Y, Shu Q, Xu J. Human adipose-derived mesenchymal stem cells alleviate obliterative bronchiolitis in a murine model via IDO. Respir Res 2017; 18:119. [PMID: 28619045 PMCID: PMC5472885 DOI: 10.1186/s12931-017-0599-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/26/2017] [Indexed: 02/08/2023] Open
Abstract
Background Long-term survival of lung transplantation is hindered by the development of obliterative bronchiolitis (OB). Adipose-derived stem cells (ASCs) were documented to have more potent immunosuppressive ability than mesenchymal stem cells (MSCs) from bone marrow and placenta. The goal of our study is to evaluate the effect of repeated administration of ASCs on OB and the involvement of indoleamine 2,3-dioxygenase (IDO) mediating the protective effect of ASCs in a heterotopic tracheal transplantation (HTT) model. Methods For studies in vitro, ASCs were treated with interferon-γ (IFN-γ). For in vivo study, tracheas from BALB/c or C57BL/6 donors were transplanted into C57BL/6 recipients to create a HTT model. On days 0, 1, 3, 5, 8, 12, 15, 20 and 25 post-transplant, the allogeneic recipient mice were administered intravenously with phosphate buffered saline, 1 × 106 human ASCs, or 1 × 106 human ASCs plus 1-methyltryptophan (1-MT), an IDO inhibitor. On days 3, 7, 14 and 28, serum, trachea and spleen samples were harvested for analysis. Results ASCs homed to heterotopic tracheal grafts after infusion. Multiple doses of ASCs significantly increased tracheal IDO levels in allografts. There were significant increases in graft and serum IFN-γ levels in allografts compared with isografts. IFN-γ elevated IDO expression and activity in ASCs in vitro. ASCs alleviated OB in allografts as evidenced by reduced epithelial loss, epithelial apoptosis, and intraluminal obstruction. The effects of ASCs on OB were blocked by 1-MT. 1-MT also blocked the alterations in pro and anti-inflammatory cytokines as well as CD3+ T cell infiltration induced by ASCs. ASCs induced not only splenic levels of CD4+CD25+Foxp3+ regulatory T cells (Treg) but also IL-10 and TGF-β-producing Treg. Furthermore, IDO inhibition abolished the changes of splenic Treg induced by ASCs. In addition, Treg reduction by cyclophosphamide treatment did not alter the effects of ASCs on tracheal IDO expression in allografts confirming Treg induction is downstream of IDO. Conclusions Repeated doses of ASCs are capable of ameliorating OB. ASCs act at least in part via elevating IDO expression. ASCs promote the generation of Treg and suppress T cell infiltration via an IDO-dependent mechanism.
Collapse
Affiliation(s)
- Guoping Zheng
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Guanguan Qiu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Menghua Ge
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Jianping He
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Lanfang Huang
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Ping Chen
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Wei Wang
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Qi Xu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China
| | - Yaoqin Hu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China
| | - Qiang Shu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China.
| | - Jianguo Xu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China. .,The First Affiliated Hospital of Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
64
|
Lopez-Santalla M, Mancheño-Corvo P, Escolano A, Menta R, DelaRosa O, Abad JL, Büscher D, Redondo JM, Bueren JA, Dalemans W, Lombardo E, Garin MI. Biodistribution and Efficacy of Human Adipose-Derived Mesenchymal Stem Cells Following Intranodal Administration in Experimental Colitis. Front Immunol 2017. [PMID: 28642759 PMCID: PMC5462906 DOI: 10.3389/fimmu.2017.00638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a large potential in cell therapy for treatment of inflammatory and autoimmune diseases, thanks to their immunomodulatory properties. The encouraging results in animal models have initiated the translation of MSC therapy to clinical trials. In cell therapy protocols with MSCs, administered intravenously, several studies have shown that a small proportion of infused MSCs can traffic to the draining lymph nodes (LNs). This is accompanied with an increase of different types of regulatory immune cells in the LNs, suggesting the importance of migration of MSCs to the LNs in order to contribute to immunomodulatory response. Intranodal (IN), also referred as intralymphatic, injection of cells, like dendritic cells, is being proposed in the clinic for the treatment of cancer and allergy, showing that this route of administration is clinically safe and efficient. In this study, we investigated, for the first time, the biodistribution and the efficacy of Luciferase+ adipose-derived MSCs (Luci-eASCs), infused through the inguinal LNs (iLNs), in normal mice and in inflamed mice with colitis. Most of the Luci-eASCs remain in the iLNs and in the adipose tissue surrounding the inguinal LNs. A small proportion of Luci-eASCs can migrate to other locations within the lymphatic system and to other tissues and organs, having a preferential migration toward the intestine in colitic mice. Our results show that the infused Luci-eASCs protected 58% of the mice against induced colitis. Importantly, a correlation between the response to eASC treatment and a higher accumulation of eASCs in popliteal, parathymic, parathyroid, and mesenteric LNs were found. Altogether, these results suggest that IN administration of eASCs is feasible and may represent an effective strategy for cell therapy protocols with human adipose-derived MSCs in the clinic for the treatment of immune-mediated disorders.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | - Amelia Escolano
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | | | | | | - Juan M Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|
65
|
Hasby Saad MA, Hasby EA. Trichinella Spiralis Impact on Mesenchymal Stem Cells: Immunohistochemical Study by Image Analyzer in Murine Model. Exp Mol Pathol 2017; 102:396-407. [PMID: 28456661 DOI: 10.1016/j.yexmp.2017.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/23/2017] [Accepted: 04/11/2017] [Indexed: 02/08/2023]
Abstract
This study aims to elucidate whether Trichinella spiralis infection or its crude antigen administration can stimulate recruitment of CD105+ve/CD45-ve cells that could represent MSCs in intestine and skeletal muscle of experimental BALB/c albino mice compared to healthy control mice. Studied mice were divided into: 20 healthy control, 20 with orally induced T. spiralis infection, 20 received adult worm crude antigen orally and 20 received larval crude antigen intramuscular. According to specific timing schedule, mice were sacrificed and tissue sections were examined for CD105 and CD45 immunohistochemical expression using image J image analyzing software, to compare different study groups. T. spiralis infection induced a significant increase in density of CD105+ve/CD45-ve cells that could represent MSCs in both intestinal and muscle sections, similarly the intramuscular injected larval crude antigen caused more infiltration of such cells in muscles compared to muscle sections from healthy control mice. However, no significant difference was noticed in intestinal sections after oral adult crude antigen administration compared to healthy control mice. So, injected T. spiralis crude antigen might be a successful stimulant to MSCs attraction and recruitment in tissues nearby injection site. This could be beneficial for cell regeneration and tissue repair in case of presence of a disease induced damage.
Collapse
Affiliation(s)
| | - Eiman A Hasby
- Pathology Department, Tanta Faculty of Medicine, Egypt.
| |
Collapse
|
66
|
Gurgul A, Opiela J, Pawlina K, Szmatoła T, Bochenek M, Bugno-Poniewierska M. The effect of histone deacetylase inhibitor trichostatin A on porcine mesenchymal stem cell transcriptome. Biochimie 2017; 139:56-73. [PMID: 28552396 DOI: 10.1016/j.biochi.2017.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/23/2017] [Indexed: 12/29/2022]
Abstract
The use of histone deacetylase inhibitors such as trichostatin A (TSA) for epigenetic transformation of mesenchymal stem cells (MSCs), whose nuclei will be transferred into enucleated oocytes, is a novel approach in research involving somatic cell cloning of pigs and other mammalian species. Although the effectiveness of TSA in cloning applications was confirmed, processes and mechanisms underlying achieved effects are not yet fully understood, especially for pig MSCs. To contribute to this knowledge, in this study we performed a comprehensive transcriptome analysis using high-throughput sequencing of pig bone-marrow derived MSCs, both treated and untreated with TSA, and evaluated the effect of TSA administration on their transcription profile after 24 h of in vitro culture. The expression of selected positive and negative mesenchymal surface antigens was also evaluated in these cells by flow cytometry. Subsequently, the stability of induced expression changes was evaluated after another 55-72 h of culture without TSA. The results of this study showed that TSA does not affect the expression of the selected surface antigens related to MSC mesenchymal stemness origin, namely: CD90 (positive marker), CD31 and CD34 (negative markers) and has a wide stimulating effect on MSCs transcription, affecting genes across the whole genome with some minor signs of site-specific acting in regions on SSC2 and SSC6. TSA turned out to have a higher impact on already expressed genes with only minor abilities to induce expression of silenced genes. Genes with expression affected by TSA were related to a wide range of biological processes, however, we found some evidence for specific stimulation of genes associated with development, differentiation, neurogenesis or myogenesis. TSA also seemed to interfere with Wnt signaling pathways by upregulation of several engaged genes. The analysis of cell transcriptome after prolonged culture following the TSA removal, showed that the expression level of majority of genes affected by TSA is restored to the initial level. Nonetheless, the set of about six hundred genes responsible for e.g. adhesion, signal transduction and cell communication was altered even after 55-72 h of culture without TSA. TSA also enhanced expression of some of pluripotency marker genes (FGF2, LIF, TERT) but their expression was stabilized during further culture without TSA. The detailed analysis of factors connected with neuron-like differentiation allowed us to assume that TSA mostly stimulates neurogenic differentiation pathway in the pig MSCs possibly through interaction with Wnt-mediated signaling and thus triggers mechanisms conducive to epigenetic reprograming.
Collapse
Affiliation(s)
- Artur Gurgul
- National Research Institute of Animal Production, Department of Genomics and Molecular Biology, Krakowska 1, 32-083, Balice, Poland.
| | - Jolanta Opiela
- National Research Institute of Animal Production, Department of Biotechnology of Animal Reproduction, Krakowska 1, 32-083, Balice, Poland
| | - Klaudia Pawlina
- National Research Institute of Animal Production, Department of Genomics and Molecular Biology, Krakowska 1, 32-083, Balice, Poland
| | - Tomasz Szmatoła
- National Research Institute of Animal Production, Department of Genomics and Molecular Biology, Krakowska 1, 32-083, Balice, Poland
| | - Michał Bochenek
- National Research Institute of Animal Production, Department of Biotechnology of Animal Reproduction, Krakowska 1, 32-083, Balice, Poland
| | - Monika Bugno-Poniewierska
- National Research Institute of Animal Production, Department of Genomics and Molecular Biology, Krakowska 1, 32-083, Balice, Poland
| |
Collapse
|
67
|
Xu C, Fu F, Li X, Zhang S. Mesenchymal stem cells maintain the microenvironment of central nervous system by regulating the polarization of macrophages/microglia after traumatic brain injury. Int J Neurosci 2017; 127:1124-1135. [PMID: 28464695 DOI: 10.1080/00207454.2017.1325884] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs), which are regarded as promising candidates for cell replacement therapies, are able to regulate immune responses after traumatic brain injury (TBI). Secondary immune response following the mechanical injury is the essential factor leading to the necrosis and apoptosis of neural cells during and after the cerebral edema has subsided and there is lack of efficient agent that can mitigate such neuroinflammation in the clinical application. By means of three molecular pathways (prostaglandin E2 (PGE2), tumor-necrosis-factor-inducible gene 6 protein (TSG-6), and progesterone receptor (PR) and glucocorticoid receptors (GR)), MSCs induce the activation of macrophages/microglia and drive them polarize into the M2 phenotypes, which inhibits the release of pro-inflammatory cytokines and promotes tissue repair and nerve regeneration. The regulation of MSCs and the polarization of macrophages/microglia are dynamically changing based on the inflammatory environment. Under the stimulation of platelet lysate (PL), MSCs also promote the release of pro-inflammatory cytokines. Meanwhile, the statue of macrophages/microglia exerts significant effects on the survival, proliferation, differentiation and activation of MSCs by changing the niche of cells. They form positive feedback loops in maintaining the homeostasis after TBI to relieving the secondary injury and promoting tissue repair. MSC therapies have obtained great achievements in several central nervous system disease clinical trials, which will accelerate the application of MSCs in TBI treatment.
Collapse
Affiliation(s)
- Chao Xu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Feng Fu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Xiaohong Li
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Sai Zhang
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| |
Collapse
|
68
|
Mancheño-Corvo P, Lopez-Santalla M, Menta R, DelaRosa O, Mulero F, Del Rio B, Ramirez C, Büscher D, Bueren JA, Lopez-Belmonte J, Dalemans W, Garin MI, Lombardo E. Intralymphatic Administration of Adipose Mesenchymal Stem Cells Reduces the Severity of Collagen-Induced Experimental Arthritis. Front Immunol 2017; 8:462. [PMID: 28484460 PMCID: PMC5399019 DOI: 10.3389/fimmu.2017.00462] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/04/2017] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with immunomodulatory properties. They have emerged as a very promising treatment for autoimmunity and inflammatory diseases such as rheumatoid arthritis. Previous studies have demonstrated that MSCs, administered systemically, migrate to lymphoid tissues associated with the inflammatory site where functional MSC-induced immune cells with a regulatory phenotype were increased mediating the immunomodulatory effects of MSCs. These results suggest that homing of MSCs to the lymphatic system plays an important role in the mechanism of action of MSCs in vivo. Thus, we hypothesized that direct intralymphatic (IL) (also referred as intranodal) administration of MSCs could be an alternative and effective route of administration for MSC-based therapy. Here, we report the feasibility and efficacy of the IL administration of human expanded adipose mesenchymal stem cells (eASCs) in a mouse model of collagen-induced arthritis (CIA). IL administration of eASCs attenuated the severity and progression of arthritis, reduced bone destruction and increased the levels of regulatory T cells (CD25+Foxp3+CD4+ cells) and Tr1 cells (IL10+CD4+), in spleen and draining lymph nodes. Taken together, these results indicate that IL administration of eASCs is very effective in modulating established CIA and may represent an alternative treatment modality for cell therapy with eASCs.
Collapse
Affiliation(s)
| | - Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Francisca Mulero
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | | | | | | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | |
Collapse
|
69
|
Klinker MW, Marklein RA, Lo Surdo JL, Wei CH, Bauer SR. Morphological features of IFN-γ-stimulated mesenchymal stromal cells predict overall immunosuppressive capacity. Proc Natl Acad Sci U S A 2017; 114:E2598-E2607. [PMID: 28283659 PMCID: PMC5380055 DOI: 10.1073/pnas.1617933114] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stromal cell (MSC) lines can vary significantly in their functional characteristics, and the effectiveness of MSC-based therapeutics may be realized by finding predictive features associated with MSC function. To identify features associated with immunosuppressive capacity in MSCs, we developed a robust in vitro assay that uses principal-component analysis to integrate multidimensional flow cytometry data into a single measurement of MSC-mediated inhibition of T-cell activation. We used this assay to correlate single-cell morphological data with overall immunosuppressive capacity in a cohort of MSC lines derived from different donors and manufacturing conditions. MSC morphology after IFN-γ stimulation significantly correlated with immunosuppressive capacity and accurately predicted the immunosuppressive capacity of MSC lines in a validation cohort. IFN-γ enhanced the immunosuppressive capacity of all MSC lines, and morphology predicted the magnitude of IFN-γ-enhanced immunosuppressive activity. Together, these data identify MSC morphology as a predictive feature of MSC immunosuppressive function.
Collapse
Affiliation(s)
- Matthew W Klinker
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Jessica L Lo Surdo
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Cheng-Hong Wei
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| |
Collapse
|
70
|
Circulating Bone Marrow-Derived CD45-/CD34+/CD133+/VEGF+ Endothelial Progenitor Cells in Adults with Crohn's Disease. Dig Dis Sci 2017; 62:633-638. [PMID: 27339637 DOI: 10.1007/s10620-016-4234-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Circulating endothelial progenitor cells (EPCs) are bone marrow-derived stem cells able to migrate to sites of damaged endothelium and differentiate into endothelial cells. Altered EPC level and function have been described in various inflammatory diseases and have been shown to augment vasculogenesis in murine models. Previous studies of EPC in the context of Crohn's disease (CD) have yielded conflicting results. AIM To determine whether the circulating levels of EPCs are changed in the context of CD. METHODS CD patients and healthy controls were recruited. Disease activity was assessed by CDAI. Peripheral blood mononuclear cells were isolated and EPC numbers evaluated by FACS analysis using anti-CD34, anti-VEGF receptor-2, anti-CD133, and anti-CD45 markers. RESULTS Eighty-three subjects, including 32 CD patients and 51 controls were recruited, including 19 (59.4 %) and 23 (45 %) males (p = 0.26), aged 34.8 ± 14.9 and 43.3 ± 18.5 years (p = 0.64), in cases and controls, respectively. Mean CDAI was 147 ± 97, disease duration was 12.7 ± 11.1 years, and 28 (87.5 %) were receiving biologics for a mean duration of 21.7 ± 16.8 months. The mean level of peripheral EPCs in CD patients was 0.050 ± 0.086 percent and 0.007 ± 0.013 % in controls (p < 0.01). There was no significant correlation between EPC levels and age (r = -0.13, p = 0.47), CDAI (r = -0.26, p = 0.15), disease duration (r = -0.04, p = 0.84), or duration of treatment with biologics (r = 0.004, p = 0.99). CONCLUSION EPCs are elevated in patients with CD. Further studies are needed to examine the function of EPCs and their possible role as a marker of disease severity or therapeutic response.
Collapse
|
71
|
Li X, Wang Y, An G, Liang D, Zhu Z, Lian X, Niu P, Guo C, Tian L. Bone marrow mesenchymal stem cells attenuate silica-induced pulmonary fibrosis via paracrine mechanisms. Toxicol Lett 2017; 270:96-107. [DOI: 10.1016/j.toxlet.2017.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/21/2022]
|
72
|
Shahini A, Mistriotis P, Asmani M, Zhao R, Andreadis ST. NANOG Restores Contractility of Mesenchymal Stem Cell-Based Senescent Microtissues. Tissue Eng Part A 2017; 23:535-545. [PMID: 28125933 DOI: 10.1089/ten.tea.2016.0494] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been extensively used in the field of tissue engineering as a source of smooth muscle cells (SMCs). However, recent studies showed deficits in the contractile function of SMCs derived from senescent MSCs and there are no available strategies to restore the contractile function that is impaired due to cellular or organismal senescence. In this study, we developed a tetracycline-regulatable system and employed micropost tissue arrays to evaluate the effects of the embryonic transcription factor, NANOG, on the contractility of senescent MSCs. Using this system, we show that expression of NANOG fortified the actin cytoskeleton and restored contractile function that was impaired in senescent MSCs. NANOG increased the expression of smooth muscle α-actin (ACTA2) as well as the contractile force generated by cells in three-dimensional microtissues. Interestingly, NANOG worked together with transforming growth factor-beta1 to further enhance the contractility of senescent microtissues. The effect of NANOG on contractile function was sustained for about 10 days after termination of its expression. Our results show that NANOG could reverse the effects of stem cell senescence and restore the myogenic differentiation potential of senescent MSCs. These findings may enable development of novel strategies to restore the function of senescent cardiovascular and other SMC-containing tissues.
Collapse
Affiliation(s)
- Aref Shahini
- 1 Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Amherst, New York
| | - Panagiotis Mistriotis
- 1 Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Amherst, New York
| | - Mohammadnabi Asmani
- 2 Department of Biomedical Engineering, University at Buffalo, The State University of New York , Amherst, New York
| | - Ruogang Zhao
- 2 Department of Biomedical Engineering, University at Buffalo, The State University of New York , Amherst, New York
| | - Stelios T Andreadis
- 1 Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York , Amherst, New York.,2 Department of Biomedical Engineering, University at Buffalo, The State University of New York , Amherst, New York.,3 Center of Excellence in Bioinformatics and Life Sciences , Buffalo, New York
| |
Collapse
|
73
|
Mesenchymal Stem Cell Administration in Patients with Chronic Obstructive Pulmonary Disease: State of the Science. Stem Cells Int 2017; 2017:8916570. [PMID: 28303154 PMCID: PMC5337878 DOI: 10.1155/2017/8916570] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/22/2017] [Indexed: 01/10/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have chronic, irreversible airway inflammation; currently, there is no effective or curative treatment and the main goals of COPD management are to mitigate symptoms and improve patients' quality of life. Stem cell based therapy offers a promising therapeutic approach that has shown potential in diverse degenerative lung diseases. Preclinical studies have demonstrated encouraging outcomes of mesenchymal stem/stromal cells (MSCs) therapy for lung disorders including emphysema, bronchopulmonary dysplasia, fibrosis, and acute respiratory distress syndrome. This review summarizes available data on 15 studies currently registered by the ClinicalTrials.gov repository, which used different stem cell therapy protocols for COPD; these included bone marrow mononuclear cells (BMMCs), bone marrow-derived MSCs, adipose-derived stem/stromal cells (ADSCs), and adipose-derived MSCs. Published results of three trials indicate that administering BMMCs or MSCs in the setting of degenerative lung disease is safe and may improve patients' condition and quality of life; however, larger-scale studies are needed to evaluate efficacy. Results of another completed trial (NCT01872624) are not yet published, and eleven other studies are ongoing; these include MSCs therapy in emphysema, several studies of ADSCs in COPD, another in idiopathic pulmonary fibrosis, and plerixafor mobilization of CD117 stem cells to peripheral blood.
Collapse
|
74
|
Ropper AE, Thakor DK, Han I, Yu D, Zeng X, Anderson JE, Aljuboori Z, Kim SW, Wang H, Sidman RL, Zafonte RD, Teng YD. Defining recovery neurobiology of injured spinal cord by synthetic matrix-assisted hMSC implantation. Proc Natl Acad Sci U S A 2017; 114:E820-E829. [PMID: 28096400 PMCID: PMC5293074 DOI: 10.1073/pnas.1616340114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal stem cells (MSCs) isolated from adult tissues offer tangible potential for regenerative medicine, given their feasibility for autologous transplantation. MSC research shows encouraging results in experimental stroke, amyotrophic lateral sclerosis, and neurotrauma models. However, further translational progress has been hampered by poor MSC graft survival, jeopardizing cellular and molecular bases for neural repair in vivo. We have devised an adult human bone marrow MSC (hMSC) delivery formula by investigating molecular events involving hMSCs incorporated in a uniquely designed poly(lactic-co-glycolic) acid scaffold, a clinically safe polymer, following inflammatory exposures in a dorsal root ganglion organotypic coculture system. Also, in rat T9-T10 hemisection spinal cord injury (SCI), we demonstrated that the tailored scaffolding maintained hMSC stemness, engraftment, and led to robust motosensory improvement, neuropathic pain and tissue damage mitigation, and myelin preservation. The scaffolded nontransdifferentiated hMSCs exerted multimodal effects of neurotrophism, angiogenesis, neurogenesis, antiautoimmunity, and antiinflammation. Hindlimb locomotion was restored by reestablished integrity of submidbrain circuits of serotonergic reticulospinal innervation at lumbar levels, the propriospinal projection network, neuromuscular junction, and central pattern generator, providing a platform for investigating molecular events underlying the repair impact of nondifferentiated hMSCs. Our approach enabled investigation of recovery neurobiology components for injured adult mammalian spinal cord that are different from those involved in normal neural function. The uncovered neural circuits and their molecular and cellular targets offer a biological underpinning for development of clinical rehabilitation therapies to treat disabilities and complications of SCI.
Collapse
Affiliation(s)
- Alexander E Ropper
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Devang K Thakor
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - InBo Han
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Dou Yu
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Xiang Zeng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Jamie E Anderson
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Zaid Aljuboori
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| | - Soo-Woo Kim
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130
| | - Hongjun Wang
- Biomedical Engineering, Chemistry, and Biological Sciences, Stevens Institute of Technology, Hoboken, NJ 07030
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School/Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Ross D Zafonte
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
| | - Yang D Teng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA 02130;
- Department of Physical Medicine & Rehabilitation, Harvard Medical School/Spaulding Rehabilitation Hospital, Charlestown, MA 02129
- Department of Neurosurgery, Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115
| |
Collapse
|
75
|
Liu Q, Zhang RZ, Li D, Cheng S, Yang YH, Tian T, Pan XR. Muse Cells, a New Type of Pluripotent Stem Cell Derived from Human Fibroblasts. Cell Reprogram 2016; 18:67-77. [PMID: 27055628 DOI: 10.1089/cell.2015.0085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A new type of mesenchymal stem cells (MSCs) that expresses stage-specific embryonic antigen 3 (SSEA-3) and the mesenchymal cell marker CD105 are known as multilineage-differentiating stress-enduring (Muse) cells. Studies have shown that stem cells in suspension cultures are more likely to generate embryoid body-like stem cell spheres and maintain an undifferentiated phenotype and pluripotency. We separated Muse cells derived from human dermal fibroblasts by long-term trypsin incubation (LTT) through suspension cultures in methylcellulose. The Muse cells obtained expressed several pluripotency markers, including Nanog, Oct4, Sox2, and SSEA-3, and could differentiate in vitro into cells of the three germ layers, such as hepatocytes (endodermal), neural cells (ectodermal) and adipocytes, and osteocytes (mesodermal cells). These cells showed a low level of DNA methylation and a high nucleo-cytoplasmic ratio. Our study provides an innovative and exciting platform for exploring the potential cell-based therapy of various human diseases using Muse cells as well as their great possibility for regenerative medicine.
Collapse
Affiliation(s)
- Qi Liu
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Ru-zhi Zhang
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Di Li
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Sai Cheng
- 2 Department of Dermatology, The First Affiliated Hospital of Bengbu Medical College , Anhui, 213003, China
| | - Yu-hua Yang
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Ting Tian
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Xiao-ru Pan
- 2 Department of Dermatology, The First Affiliated Hospital of Bengbu Medical College , Anhui, 213003, China
| |
Collapse
|
76
|
Wang LT, Ting CH, Yen ML, Liu KJ, Sytwu HK, Wu KK, Yen BL. Human mesenchymal stem cells (MSCs) for treatment towards immune- and inflammation-mediated diseases: review of current clinical trials. J Biomed Sci 2016; 23:76. [PMID: 27809910 PMCID: PMC5095977 DOI: 10.1186/s12929-016-0289-5] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) are multilineage somatic progenitor/stem cells that have been shown to possess immunomodulatory properties in recent years. Initially met with much skepticism, MSC immunomodulation has now been well reproduced across tissue sources and species to be clinically relevant. This has opened up the use of these versatile cells for application as 3rd party/allogeneic use in cell replacement/tissue regeneration, as well as for immune- and inflammation-mediated disease entities. Most surprisingly, use of MSCs for in immune-/inflammation-mediated diseases appears to yield more efficacy than for regenerative medicine, since engraftment of the exogenous cell does not appear necessary. In this review, we focus on this non-traditional clinical use of a tissue-specific stem cell, and highlight important findings and trends in this exciting area of stem cell therapy.
Collapse
Affiliation(s)
- Li-Tzu Wang
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan
| | - Chiao-Hsuan Ting
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan
| | - Men-Luh Yen
- Department of Ob/Gyn, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Tainan, Taiwan
| | - Huey-Kang Sytwu
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan.,Graduate Institute of Microbiology and Immunology, NDMC, Taipei, Taiwan
| | - Kenneth K Wu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.,Graduate Institute of Basic Medical Sciences, China Medical College, Taichung, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.
| |
Collapse
|
77
|
Lazennec G, Lam PY. Recent discoveries concerning the tumor - mesenchymal stem cell interactions. Biochim Biophys Acta Rev Cancer 2016; 1866:290-299. [PMID: 27750042 DOI: 10.1016/j.bbcan.2016.10.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/09/2016] [Accepted: 10/13/2016] [Indexed: 12/18/2022]
Abstract
Tumor microenvironment plays a crucial role in coordination with cancer cells in the establishment, growth and dissemination of the tumor. Among cells of the microenvironment, mesenchymal stem cells (MSCs) and their ability to evolve into cancer associated fibroblasts (CAFs) have recently generated a major interest in the field. Numerous studies have described the potential pro- or anti-tumorigenic action of MSCs. The goal of this review is to synthesize recent and emerging discoveries concerning the mechanisms by which MSCs can be attracted to tumor sites, how they can generate CAFs and by which way MSCs are able to modulate the growth, response to treatments, angiogenesis, invasion and metastasis of tumors. The understanding of the role of MSCs in tumor development has potential and clinical applications in terms of cancer management.
Collapse
Affiliation(s)
- Gwendal Lazennec
- CNRS, SYS2DIAG, Cap delta, 1682 rue de la Valsière, Montpellier F-34184, France; CNRS, GDR 3697 "Microenvrionment of tumor niches", Micronit, France.
| | - Paula Y Lam
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Cancer and Stem Cells Biology Program, Duke-NUS Graduate Medical School, Singapore 169857, Singapore.
| |
Collapse
|
78
|
Varshney R, Ali Q, Wu C, Sun Z. Monocrotaline-Induced Pulmonary Hypertension Involves Downregulation of Antiaging Protein Klotho and eNOS Activity. Hypertension 2016; 68:1255-1263. [PMID: 27672025 DOI: 10.1161/hypertensionaha.116.08184] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022]
Abstract
The objective of this study is to investigate whether stem cell delivery of secreted Klotho (SKL), an aging-suppressor protein, attenuates monocrotaline-induced pulmonary vascular dysfunction and remodeling. Overexpression of SKL in mesenchymal stem cells (MSCs) was achieved by transfecting MSCs with lentiviral vectors expressing SKL-green fluorescent protein (GFP). Four groups of rats were treated with monocrotaline, whereas an additional group was given saline (control). Three days later, 4 monocrotaline-treated groups received intravenous delivery of nontransfected MSCs, MSC-GFP, MSC-SKL-GFP, and PBS, respectively. Ex vivo vascular relaxing responses to acetylcholine were diminished in small pulmonary arteries (PAs) in monocrotaline-treated rats, indicating pulmonary vascular endothelial dysfunction. Interestingly, delivery of MSCs overexpressing SKL (MSC-SKL-GFP) abolished monocrotaline-induced pulmonary vascular endothelial dysfunction and PA remodeling. Monocrotaline significantly increased right ventricular systolic blood pressure, which was attenuated significantly by MSC-SKL-GFP, indicating improved PA hypertension. MSC-SKL-GFP also attenuated right ventricular hypertrophy. Nontransfected MSCs slightly, but not significantly, improved PA hypertension and pulmonary vascular endothelial dysfunction. MSC-SKL-GFP attenuated monocrotaline-induced inflammation, as evidenced by decreased macrophage infiltration around PAs. MSC-SKL-GFP increased SKL levels, which rescued the downregulation of SIRT1 (Sirtuin 1) expression and endothelial NO synthase (eNOS) phosphorylation in the lungs of monocrotaline-treated rats. In cultured endothelial cells, SKL abolished monocrotaline-induced downregulation of eNOS activity and NO levels and enhanced cell viability. Therefore, stem cell delivery of SKL is an effective therapeutic strategy for pulmonary vascular endothelial dysfunction and PA remodeling. SKL attenuates monocrotaline-induced PA remodeling and PA smooth muscle cell proliferation, likely by reducing inflammation and restoring SIRT1 levels and eNOS activity.
Collapse
Affiliation(s)
- Rohan Varshney
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Quaisar Ali
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Chengxiang Wu
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
79
|
Nikiforou M, Willburger C, de Jong AE, Kloosterboer N, Jellema RK, Ophelders DRMG, Steinbusch HWM, Kramer BW, Wolfs TGAM. Global hypoxia-ischemia induced inflammation and structural changes in the preterm ovine gut which were not ameliorated by mesenchymal stem cell treatment. Mol Med 2016; 22:244-257. [PMID: 27257938 PMCID: PMC5023518 DOI: 10.2119/molmed.2015.00252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
Perinatal asphyxia, a condition of impaired gas exchange during birth, leads to fetal hypoxia-ischemia (HI) and is associated with postnatal adverse outcomes including intestinal dysmotility and necrotizing enterocolitis (NEC). Evidence from adult animal models of transient, locally-induced intestinal HI has shown that inflammation is essential in HI-induced injury of the gut. Importantly, mesenchymal stem cell (MSC) treatment prevented this HI-induced intestinal damage. We therefore assessed whether fetal global HI induced inflammation, injury and developmental changes in the gut and whether intravenous MSC administration ameliorated these HI-induced adverse intestinal effects. In a preclinical ovine model, fetuses were subjected to umbilical cord occlusion (UCO), with or without MSC treatment, and sacrificed 7 days after UCO. Global HI increased the number of myeloperoxidase positive cells in the mucosa, upregulated mRNA levels of interleukin (IL)-1β and IL-17 in gut tissue and caused T-cell invasion in the intestinal muscle layer. Intestinal inflammation following global HI was associated with increased Ki67+ cells in the muscularis and subsequent muscle hyperplasia. Global HI caused distortion of glial fibrillary acidic protein immunoreactivity in the enteric glial cells and increased synaptophysin and serotonin expression in the myenteric ganglia. Intravenous MSC treatment did not ameliorate these HI-induced adverse intestinal events. Global HI resulted in intestinal inflammation and enteric nervous system abnormalities which are clinically associated with postnatal complications including feeding intolerance, altered gastrointestinal transit and NEC. The intestinal histopathological changes were not prevented by intravenous MSC treatment directly after HI, indicating that alternative treatment regimens for cell-based therapies should be explored.
Collapse
Affiliation(s)
- Maria Nikiforou
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Carolin Willburger
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anja E de Jong
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nico Kloosterboer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daan RMG Ophelders
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Harry WM Steinbusch
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim GAM Wolfs
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
80
|
Little MH, Kairath P. Regenerative medicine in kidney disease. Kidney Int 2016; 90:289-299. [DOI: 10.1016/j.kint.2016.03.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/10/2016] [Accepted: 03/17/2016] [Indexed: 12/31/2022]
|
81
|
Ribeiro A, Ritter T, Griffin M, Ceredig R. Development of a flow cytometry-based potency assay for measuring the in vitro immunomodulatory properties of mesenchymal stromal cells. Immunol Lett 2016; 177:38-46. [PMID: 27451032 DOI: 10.1016/j.imlet.2016.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/04/2016] [Accepted: 07/19/2016] [Indexed: 01/14/2023]
Abstract
Human bone marrow-derived mesenchymal stromal/stem cells (MSC) have well-documented modulatory effects on multiple immune cell types. Although these effects are linked to their therapeutic benefit in diverse diseases, a reliable, quantitative assay of the immunomodulatory potency of individual human MSC preparations is lacking. The aims of this study were to develop an optimised rapid turnaround, flow cytometry-based whole-blood assay to monitor MSC potency and to validate its application to MSC immunomodulation. A protocol for short-term LPS stimulation of anti-coagulated whole blood samples followed by combined surface CD45/CD14 and intracellular TNF-α staining was initially developed for analysis on a 4 colour desktop cytometer. Optimal monocyte activation was dependent on the presence of extracellular calcium ions thereby precluding the use of EDTA and sodium citrate as anticoagulants. Optimal assay conditions proved to be 1ng/mL ultrapure-LPS added to 10-fold diluted, heparin anti-coagulated whole blood incubated for 6h at 37°C. Under these conditions, addition of human bone marrow-derived MSC (hBM-MSC) from multiple donors resulted in a reproducible, dose-dependent inhibition of LPS-stimulated monocyte TNF-α expression. We conclude that this protocol represents a practical, quantitative assay of a clinically relevant functional effect of hBM-MSCs as well as other immunomodulatory agents.
Collapse
Affiliation(s)
- Andreia Ribeiro
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| | - Thomas Ritter
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| | - Matthew Griffin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| | - Rhodri Ceredig
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and School of Medicine, National University of Ireland, Galway, Ireland.
| |
Collapse
|
82
|
Dykstra B, Lee J, Mortensen LJ, Yu H, Wu ZL, Lin CP, Rossi DJ, Sackstein R. Glycoengineering of E-Selectin Ligands by Intracellular versus Extracellular Fucosylation Differentially Affects Osteotropism of Human Mesenchymal Stem Cells. Stem Cells 2016; 34:2501-2511. [PMID: 27335219 DOI: 10.1002/stem.2435] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022]
Abstract
Human mesenchymal stem cells (MSCs) hold great promise in cellular therapeutics for skeletal diseases but lack expression of E-selectin ligands that direct homing of blood-borne cells to bone marrow. Previously, we described a method to engineer E-selectin ligands on the MSC surface by exofucosylating cells with fucosyltransferase VI (FTVI) and its donor sugar, GDP-Fucose, enforcing transient surface expression of the potent E-selectin ligand HCELL with resultant enhanced osteotropism of intravenously administered cells. Here, we sought to determine whether E-selectin ligands created via FTVI-exofucosylation are distinct in identity and function to those created by FTVI expressed intracellularly. To this end, we introduced synthetic modified mRNA encoding FTVI (FUT6-modRNA) into human MSCs. FTVI-exofucosylation (i.e., extracellular fucosylation) and FUT6-modRNA transfection (i.e., intracellular fucosylation) produced similar peak increases in cell surface E-selectin ligand levels, and shear-based functional assays showed comparable increases in tethering/rolling on human endothelial cells expressing E-selectin. However, biochemical analyses revealed that intracellular fucosylation induced expression of both intracellular and cell surface E-selectin ligands and also induced a more sustained expression of E-selectin ligands compared to extracellular fucosylation. Notably, live imaging studies to assess homing of human MSC to mouse calvarium revealed more osteotropism following intravenous administration of intracellularly-fucosylated cells compared to extracellularly-fucosylated cells. This study represents the first direct analysis of E-selectin ligand expression programmed on human MSCs by FTVI-mediated intracellular versus extracellular fucosylation. The observed differential biologic effects of FTVI activity in these two contexts may yield new strategies for improving the efficacy of human MSCs in clinical applications. Stem Cells 2016;34:2501-2511.
Collapse
Affiliation(s)
- Brad Dykstra
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard University, Cambridge, Massachusetts, USA.,Program of Excellence in Glycosciences, Harvard University, Cambridge, Massachusetts, USA
| | - Jungmin Lee
- Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Harvard University, Cambridge, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Luke J Mortensen
- Regenerative Bioscience Center, Rhodes Center for Animal and Dairy Science and College of Engineering, University of Georgia, Athens, Georgia, USA
| | - Haixiao Yu
- Bio-Techne, R&D Systems, Inc, Minneapolis, Massachusetts, USA
| | - Zhengliang L Wu
- Bio-Techne, R&D Systems, Inc, Minneapolis, Massachusetts, USA
| | - Charles P Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Derrick J Rossi
- Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Harvard University, Cambridge, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Robert Sackstein
- Department of Dermatology and Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard University, Cambridge, Massachusetts, USA. .,Program of Excellence in Glycosciences, Harvard University, Cambridge, Massachusetts, USA. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
83
|
Griffin TP, Martin WP, Islam N, O'Brien T, Griffin MD. The Promise of Mesenchymal Stem Cell Therapy for Diabetic Kidney Disease. Curr Diab Rep 2016; 16:42. [PMID: 27007719 DOI: 10.1007/s11892-016-0734-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus (DM) commonly leads to progressive chronic kidney disease despite current best medical practice. The pathogenesis of diabetic kidney disease (DKD) involves a complex network of primary and secondary mechanisms with both intra-renal and systemic components. Apart from inhibition of the renin angiotensin aldosterone system, targeting individual pathogenic mediators with drug therapy has not, thus far, been proven to have high clinical value. Stem or progenitor cell therapies offer an alternative strategy for modulating complex disease processes through suppressing multiple pathogenic pathways and promoting pro-regenerative mechanisms. Mesenchymal stem cells (MSCs) have shown particular promise based on their accessibility from adult tissues and their diverse mechanisms of action including secretion of paracrine anti-inflammatory and cyto-protective factors. In this review, the progress toward clinical translation of MSC therapy for DKD is critically evaluated. Results from animal models suggest distinct potential for systemic MSC infusion to favourably modulate DKD progression. However, only a few early phase clinical trials have been initiated and efficacy in humans remains to be proven. Key knowledge gaps and research opportunities exist in this field. These include the need to gain greater understanding of in vivo mechanism of action, to identify quantifiable biomarkers of response to therapy and to define the optimal source, dose and timing of MSC administration. Given the rising prevalence of DM and DKD worldwide, continued progress toward harnessing the inherent regenerative functions of MSCs and other progenitor cells for even a subset of those affected has potential for profound societal benefits.
Collapse
Affiliation(s)
- Tomás P Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
- Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| | - William Patrick Martin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
- Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| | - Nahidul Islam
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
- Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.
- Nephrology Services, Galway University Hospitals, Saolta University Health Group, Galway, Ireland.
| |
Collapse
|
84
|
Khedoe PPSJ, Rensen PCN, Berbée JFP, Hiemstra PS. Murine models of cardiovascular comorbidity in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1011-27. [PMID: 26993520 DOI: 10.1152/ajplung.00013.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/15/2016] [Indexed: 01/12/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have an increased risk for cardiovascular disease (CVD). Currently, COPD patients with atherosclerosis (i.e., the most important underlying cause of CVD) receive COPD therapy complemented with standard CVD therapy. This may, however, not be the most optimal treatment. To investigate the link between COPD and atherosclerosis and to develop specific therapeutic strategies for COPD patients with atherosclerosis, a substantial number of preclinical studies using murine models have been performed. In this review, we summarize the currently used murine models of COPD and atherosclerosis, both individually and combined, and discuss the relevance of these models for studying the pathogenesis and development of new treatments for COPD patients with atherosclerosis. Murine and clinical studies have provided complementary information showing a prominent role for systemic inflammation and oxidative stress in the link between COPD and atherosclerosis. These and other studies showed that murine models for COPD and atherosclerosis are useful tools and can provide important insights relevant to understanding the link between COPD and CVD. More importantly, murine studies provide good platforms for studying the potential of promising (new) therapeutic strategies for COPD patients with CVD.
Collapse
Affiliation(s)
- P Padmini S J Khedoe
- Department of Pulmonology, Leiden University Medical Center, the Netherlands; Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, the Netherlands
| |
Collapse
|
85
|
Gazdic M, Volarevic V, Arsenijevic N, Stojkovic M. Mesenchymal stem cells: a friend or foe in immune-mediated diseases. Stem Cell Rev Rep 2016; 11:280-7. [PMID: 25592610 DOI: 10.1007/s12015-014-9583-3] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult, self-renewable, multipotent cells that can be found in almost all postnatal tissues. Because of their capacity for self-renewal and differentiation into tissues of mesodermal origin and due to their immunomodulatory ability, MSCs are used in many preclinical and clinical studies as possible new therapeutic agents for the autoimmune or degenerative diseases treatment. In dependence of inflammatory environment to which they are exposed to, MSCs adopt immunosuppressive or pro-inflammatory phenotype. In the presence of high levels of pro-inflammatory cytokines or through activation of Toll-like receptor (TLR)-3, MSCs adopt an immune-suppressive phenotype and suppress the proliferation, activation and effector function of professional antigen presenting cells (dendritic cells, macrophages, B lymphocytes), T lymphocytes, NK cells, NKT cells, and neutrophils. During the early phase of inflammation, through TLR4 activation and in the presence of low levels of inflammatory cytokines, MSCs adopt a pro-inflammatory phenotype, promote neutrophil and T cell activation and enhance immune response. Here we review the current findings on the immunoregulatory plasticity of MSCs involved in regulation of immune response.
Collapse
Affiliation(s)
- Marina Gazdic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000, Kragujevac, Serbia
| | | | | | | |
Collapse
|
86
|
Yang Y, Pang D, Hu C, Lv Y, He T, An Y, Tang Z, Deng Z. Nestin Positive Bone Marrow Derived Cells Responded to Injury Mobilize into Peripheral Circulation and Participate in Skin Defect Healing. PLoS One 2015; 10:e0143368. [PMID: 26633897 PMCID: PMC4669078 DOI: 10.1371/journal.pone.0143368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022] Open
Abstract
Exogenously infused mesenchymal stem cells (MSCs) are thought to migrate to injury site through peripheral blood stream and participate in tissue repair. However, whether and how endogenous bone marrow MSCs mobilized to circulating and targeted to tissue injury has raised some controversy, and related studies were restricted by the difficulty of MSCs identifying in vivo. Nestin, a kind of intermediate filament protein initially identified in neuroepithelial stem cells, was recently reported as a credible criteria for MSCs in bone marrow. In this study, we used a green fluorescent protein (GFP) labeled bone marrow replacement model to trace the nestin positive bone marrow derived cells (BMDCs) of skin defected-mice. We found that after skin injured, numbers of nestin+ cells in peripheral blood and bone marrow both increased. A remarkable concentration of nestin+ BMDCs around skin wound was detected, while few of these cells could be observed in uninjured skin or other organs. This recruitment effect could not be promoted by granulocyte colony-stimulating factor (G-CSF), suggests a different mobilization mechanism from ones G-CSF takes effect on hematopoietic cells. Our results proposed nestin+ BMDCs as mobilized candidates in skin injury repair, which provide a new insight of endogenous MSCs therapy.
Collapse
Affiliation(s)
- Yi Yang
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Oral Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Danlin Pang
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xiangya Stomatology Hospital, Central South University, Changsha, Hunan, China
| | - Chenghu Hu
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xi’an Institute of Tissue Engineering & Regenerative Medicine, Shaanxi, China
| | - Yajie Lv
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shannxi, China
| | - Tao He
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yulin An
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Zhangui Tang
- Xiangya Stomatology Hospital, Central South University, Changsha, Hunan, China
- * E-mail: (ZD)’ (ZT)
| | - Zhihong Deng
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- * E-mail: (ZD)’ (ZT)
| |
Collapse
|
87
|
Bhat V, Olmer M, Joshi S, Durden DL, Cramer TJ, Barnes RFW, Ball ST, Hughes TH, Silva M, Luck JV, Moore RE, Mosnier LO, von Drygalski A. Vascular remodeling underlies rebleeding in hemophilic arthropathy. Am J Hematol 2015; 90:1027-35. [PMID: 26257191 DOI: 10.1002/ajh.24133] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/10/2023]
Abstract
Hemophilic arthropathy is a debilitating condition that can develop as a consequence of frequent joint bleeding despite adequate clotting factor replacement. The mechanisms leading to repeated spontaneous bleeding are unknown. We investigated synovial, vascular, stromal, and cartilage changes in response to a single induced hemarthrosis in the FVIII-deficient mouse. We found soft-tissue hyperproliferation with marked induction of neoangiogenesis and evolving abnormal vascular architecture. While soft-tissue changes were rapidly reversible, abnormal vascularity persisted for months and, surprisingly, was also seen in uninjured joints. Vascular changes in FVIII-deficient mice involved pronounced remodeling with expression of α-Smooth Muscle Actin (SMA), Endoglin (CD105), and vascular endothelial growth factor, as well as alterations of joint perfusion as determined by in vivo imaging. Vascular architecture changes and pronounced expression of α-SMA appeared unique to hemophilia, as these were not found in joint tissue obtained from mouse models of rheumatoid arthritis and osteoarthritis and from patients with the same conditions. Evidence that vascular changes in hemophilia were significantly associated with bleeding and joint deterioration was obtained prospectively by dynamic in vivo imaging with musculoskeletal ultrasound and power Doppler of 156 joints (elbows, knees, and ankles) in a cohort of 26 patients with hemophilia at baseline and during painful episodes. These observations support the hypothesis that vascular remodeling contributes significantly to bleed propagation and development of hemophilic arthropathy. Based on these findings, the development of molecular targets for angiogenesis inhibition may be considered in this disease.
Collapse
Affiliation(s)
- Vikas Bhat
- Department of Molecular and Experimental Medicine; The Scripps Research Institute; La Jolla California
- Department of Medicine; University of California San Diego; San Diego California
| | - Merissa Olmer
- Department of Molecular and Experimental Medicine; The Scripps Research Institute; La Jolla California
| | - Shweta Joshi
- Department of Pediatrics; University of California San Diego; California
| | - Donald L. Durden
- Department of Pediatrics; University of California San Diego; California
| | - Thomas J. Cramer
- Department of Medicine; University of California San Diego; San Diego California
| | - Richard FW Barnes
- Department of Medicine; University of California San Diego; San Diego California
| | - Scott T. Ball
- Department of Orthopaedic Surgery; University of California San Diego; San Diego California
| | - Tudor H. Hughes
- Department of Radiology; University of California San Diego; San Diego California
| | - Mauricio Silva
- Orthopedic Institute for Children University of California Los Angeles; Los Angeles California
| | - James V. Luck
- Orthopedic Institute for Children University of California Los Angeles; Los Angeles California
| | | | - Laurent O. Mosnier
- Department of Molecular and Experimental Medicine; The Scripps Research Institute; La Jolla California
| | - Annette von Drygalski
- Department of Molecular and Experimental Medicine; The Scripps Research Institute; La Jolla California
- Department of Medicine; University of California San Diego; San Diego California
| |
Collapse
|
88
|
von Dalowski F, Kramer M, Wermke M, Wehner R, Röllig C, Alakel N, Stölzel F, Parmentier S, Sockel K, Krech M, Schmitz M, Platzbecker U, Schetelig J, Bornhäuser M, von Bonin M. Mesenchymal Stromal Cells for Treatment of Acute Steroid-Refractory Graft Versus Host Disease: Clinical Responses and Long-Term Outcome. Stem Cells 2015; 34:357-66. [DOI: 10.1002/stem.2224] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/31/2015] [Accepted: 09/14/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Felix von Dalowski
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Michael Kramer
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Martin Wermke
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
- Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Rebekka Wehner
- Institut für Immunologie; Technische Universität Dresden; Germany
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Nael Alakel
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Friedrich Stölzel
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Stefani Parmentier
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Rems-Murr-Klinik; Waiblingen Germany
| | - Katja Sockel
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Mathias Krech
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Marc Schmitz
- Institut für Immunologie; Technische Universität Dresden; Germany
- Deutsches Konsortium für Translationale Krebsforschung; Standort Dresden Germany
- Deutsches Krebsforschungszentrum; Heidelberg Germany
- Center for Regenerative Therapies Dresden; Dresden Germany
| | - Uwe Platzbecker
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Johannes Schetelig
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
- Deutsches Konsortium für Translationale Krebsforschung; Standort Dresden Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
- Deutsches Konsortium für Translationale Krebsforschung; Standort Dresden Germany
- Center for Regenerative Therapies Dresden; Dresden Germany
| | - Malte von Bonin
- Medizinische Klinik und Poliklinik I; Universitäts KrebsCentrum, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
- Deutsches Konsortium für Translationale Krebsforschung; Standort Dresden Germany
- Deutsches Krebsforschungszentrum; Heidelberg Germany
| |
Collapse
|
89
|
Campos AM, Maciel E, Moreira ASP, Sousa B, Melo T, Domingues P, Curado L, Antunes B, Domingues MRM, Santos F. Lipidomics of Mesenchymal Stromal Cells: Understanding the Adaptation of Phospholipid Profile in Response to Pro-Inflammatory Cytokines. J Cell Physiol 2015; 231:1024-32. [DOI: 10.1002/jcp.25191] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/10/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Ana Margarida Campos
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Elisabete Maciel
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Ana S. P. Moreira
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Bebiana Sousa
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Tânia Melo
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Liliana Curado
- Cell2B Advanced Therapeutics SA; Biocant Park Núcleo 04 Lote 4 A; Cantanhede Portugal
| | - Brígida Antunes
- Cell2B Advanced Therapeutics SA; Biocant Park Núcleo 04 Lote 4 A; Cantanhede Portugal
| | - M. Rosário M. Domingues
- Mass Spectrometry Centre, QOPNA; Department of Chemistry; University of Aveiro; Aveiro Portugal
| | - Francisco Santos
- Cell2B Advanced Therapeutics SA; Biocant Park Núcleo 04 Lote 4 A; Cantanhede Portugal
| |
Collapse
|
90
|
Kim HJ, Kwon YR, Bae YJ, Kim YJ. Enhancement of human mesenchymal stem cell differentiation by combination treatment with 5-azacytidine and trichostatin A. Biotechnol Lett 2015; 38:167-74. [PMID: 26341652 DOI: 10.1007/s10529-015-1949-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/27/2015] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To enhance the differentiation of mesenchymal stem cells (MSCs) and their epigenetic status by modification using hypomethylating agents (HMAs) and histone deacetylase inhibitors (HDACs). RESULTS Treatment with 5-azacytidine or 5-azacytidine plus trichostatin A (TSA) increased expression of Runx-2, BDNF and Sox-9 compared with the control or TSA groups. Maximal increases of 4.1-, 4.5-, and 8.3-fold in Runx-2, BDNF, and Sox-9 transcript levels, respectively, were observed in the 5-azacytidine plus TSA group. Similar to the expression pattern of key regulatory molecules, differentiation to each lineage was also enhanced considerably in the 5-azacytidine or in the 5-azacytidine plus TSA groups. Quantitative analyses at the protein level showed 8.9-, 26.8-, 27.9-, and 28.5-fold upregulation of osterix, MAP-2, nestin, and type II collagen), respectively. CONCLUSION HMAs and HDACs enhanced in vitro differentiation of MSCs, which was maximized when the two drugs were combined, with HMA having the dominant effect.
Collapse
Affiliation(s)
- Hye Joung Kim
- Laboratory of Hematological Disease and Transplant Immunology, Seoul, Republic of Korea
| | - Yong-Rim Kwon
- Laboratory of Hematological Disease and Transplant Immunology, Seoul, Republic of Korea
| | - Yu-Jin Bae
- Laboratory of Hematological Disease and Transplant Immunology, Seoul, Republic of Korea
| | - Yoo-Jin Kim
- Laboratory of Hematological Disease and Transplant Immunology, Seoul, Republic of Korea. .,Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, Catholic Blood and Marrow Transplantation Center, Catholic University Of Korea, 505 Banpo-Dong, Seocho-Gu, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
91
|
Human Adipose-Derived Mesenchymal Stem Cells Modulate Experimental Autoimmune Arthritis by Modifying Early Adaptive T Cell Responses. Stem Cells 2015. [DOI: 10.1002/stem.2113] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
92
|
Mesenchymal Stromal Cells Affect Disease Outcomes via Macrophage Polarization. Stem Cells Int 2015; 2015:989473. [PMID: 26257791 PMCID: PMC4518189 DOI: 10.1155/2015/989473] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/30/2015] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewable cells that reside in almost all postnatal tissues. In recent years, many studies have reported the effect of MSCs on the innate and adaptive immune systems. MSCs regulate the proliferation, activation, and effector function of T lymphocytes, professional antigen presenting cells (dendritic cells, macrophages, and B lymphocytes), and NK cells via direct cell-to-cell contact or production of soluble factors including indoleamine 2,3-dioxygenase, prostaglandin E2, tumor necrosis factor-α stimulated gene/protein 6, nitric oxide, and IL-10. MSCs are also able to reprogram macrophages from a proinflammatory M1 phenotype toward an anti-inflammatory M2 phenotype capable of regulating immune response. Because of their capacity for differentiation and immunomodulation, MSCs have been used in many preclinical and clinical studies as possible new therapeutic agents for the treatment of autoimmune, degenerative, and inflammatory diseases. In this review, we discuss the central role of MSCs in macrophage polarization and outcomes of diseases such as wound healing, brain/spinal cord injuries, and diseases of heart, lung, and kidney in animal models.
Collapse
|
93
|
Violatto MB, Santangelo C, Capelli C, Frapolli R, Ferrari R, Sitia L, Tortarolo M, Talamini L, Previdi S, Moscatelli D, Salmona M, Introna M, Bendotti C, Bigini P. Longitudinal tracking of triple labeled umbilical cord derived mesenchymal stromal cells in a mouse model of Amyotrophic Lateral Sclerosis. Stem Cell Res 2015; 15:243-53. [PMID: 26177481 DOI: 10.1016/j.scr.2015.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/25/2015] [Accepted: 06/26/2015] [Indexed: 12/13/2022] Open
Abstract
The translational potential of cell therapy to humans requires a deep knowledge of the interaction between transplanted cells and host tissues. In this study, we evaluate the behavior of umbilical cord mesenchymal stromal cells (UC-MSCs), labeled with fluorescent nanoparticles, transplanted in healthy or early symptomatic transgenic SOD1G93A mice (a murine model of Amyotrophic Lateral Sclerosis). The double labeling of cells with nanoparticles and Hoechst-33258 enabled their tracking for a long time in both cells and tissues. Whole-body distribution of UC-MSCs was performed by in-vivo and ex-vivo analyses 1, 7, 21 days after single intravenous or intracerebroventricular administration. By intravenous administration cells were sequestered by the lungs and rapidly cleared by the liver. No difference in biodistribution was found among the two groups. On the other hand, UC-MSCs transplanted in lateral ventricles remained on the choroid plexus for the whole duration of the study even if decreasing in number. Few cells were found in the spinal cord of SOD1G93A mice exclusively. No migration in brain parenchyma was observed. These results suggest that the direct implantation in brain ventricles allows a prolonged permanence of cells close to the damaged areas and makes this method of tracking reliable for future studies of efficacy.
Collapse
Affiliation(s)
| | - Chiara Santangelo
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Chiara Capelli
- USS Centro di Terapia Cellulare "G. Lanzani", A. O. Papa Giovanni XXIII, Bergamo, Italy
| | - Roberta Frapolli
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Raffaele Ferrari
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Leopoldo Sitia
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Massimo Tortarolo
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Laura Talamini
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Sara Previdi
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Davide Moscatelli
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "G. Natta", Politecnico di Milano, Milano, Italy
| | - Mario Salmona
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Martino Introna
- USS Centro di Terapia Cellulare "G. Lanzani", A. O. Papa Giovanni XXIII, Bergamo, Italy
| | - Caterina Bendotti
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Paolo Bigini
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| |
Collapse
|
94
|
Interleukin-17 and its implication in the regulation of differentiation and function of hematopoietic and mesenchymal stem cells. Mediators Inflamm 2015; 2015:470458. [PMID: 25999667 PMCID: PMC4427009 DOI: 10.1155/2015/470458] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells have a great potential applicability in regenerative medicine and cell-based therapies. However, there are still many unresolved issues concerning their biology, and the influence of the local microenvironment on properties of stem cells has been increasingly recognized. Interleukin (IL-) 17, as a cytokine implicated in many physiological and pathological processes, should be taken into consideration as a part of a regulatory network governing tissue-associated stem cells' fate. This review is focusing on the published data on the effects of IL-17 on the properties and function of hematopoietic and mesenchymal stem cells and trying to discuss that IL-17 achieves many of its roles by acting on adult stem cells.
Collapse
|
95
|
Klinker MW, Wei CH. Mesenchymal stem cells in the treatment of inflammatory and autoimmune diseases in experimental animal models. World J Stem Cells 2015; 7:556-567. [PMID: 25914763 PMCID: PMC4404391 DOI: 10.4252/wjsc.v7.i3.556] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/07/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Multipotent mesenchymal stromal cells [also known as mesenchymal stem cells (MSCs)] are currently being studied as a cell-based treatment for inflammatory disorders. Experimental animal models of human immune-mediated diseases have been instrumental in establishing their immunosuppressive properties. In this review, we summarize recent studies examining the effectiveness of MSCs as immunotherapy in several widely-studied animal models, including type 1 diabetes, experimental autoimmune arthritis, experimental autoimmune encephalomyelitis, inflammatory bowel disease, graft-vs-host disease, and systemic lupus erythematosus. In addition, we discuss mechanisms identified by which MSCs mediate immune suppression in specific disease models, and potential sources of functional variability of MSCs between studies.
Collapse
|
96
|
Moll G, Le Blanc K. Engineering more efficient multipotent mesenchymal stromal (stem) cells for systemic delivery as cellular therapy. ACTA ACUST UNITED AC 2015. [DOI: 10.1111/voxs.12133] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- G. Moll
- Division of Clinical Immunology and Transfusion Medicine; Department of Laboratory Medicine; Karolinska Institutet; Stockholm Sweden
- Hematology and Regenerative Medicine Centre at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - K. Le Blanc
- Division of Clinical Immunology and Transfusion Medicine; Department of Laboratory Medicine; Karolinska Institutet; Stockholm Sweden
- Hematology and Regenerative Medicine Centre at Karolinska University Hospital Huddinge; Stockholm Sweden
| |
Collapse
|
97
|
McCullagh KJA, Perlingeiro RCR. Coaxing stem cells for skeletal muscle repair. Adv Drug Deliv Rev 2015; 84:198-207. [PMID: 25049085 PMCID: PMC4295015 DOI: 10.1016/j.addr.2014.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/19/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023]
Abstract
Skeletal muscle has a tremendous ability to regenerate, attributed to a well-defined population of muscle stem cells called satellite cells. However, this ability to regenerate diminishes with age and can also be dramatically affected by multiple types of muscle diseases, or injury. Extrinsic and/or intrinsic defects in the regulation of satellite cells are considered to be major determinants for the diminished regenerative capacity. Maintenance and replenishment of the satellite cell pool is one focus for muscle regenerative medicine, which will be discussed. There are other sources of progenitor cells with myogenic capacity, which may also support skeletal muscle repair. However, all of these myogenic cell populations have inherent difficulties and challenges in maintaining or coaxing their derivation for therapeutic purpose. This review will highlight recent reported attributes of these cells and new bioengineering approaches to creating a supply of myogenic stem cells or implants applicable for acute and/or chronic muscle disorders.
Collapse
Affiliation(s)
- Karl J A McCullagh
- Department of Physiology, School of Medicine and Regenerative Medicine Institute, National University of Ireland Galway, Ireland
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
98
|
Flores AI, Gómez-Gómez GJ, Masedo-González &A, Martínez-Montiel MP. Stem cell therapy in inflammatory bowel disease: A promising therapeutic strategy? World J Stem Cells 2015; 7:343-351. [PMID: 25815119 PMCID: PMC4369491 DOI: 10.4252/wjsc.v7.i2.343] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/24/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases are inflammatory, chronic and progressive diseases of the intestinal tract for which no curative treatment is available. Research in other fields with stem cells of different sources and with immunoregulatory cells (regulatory T-lymphocytes and dendritic T-cells) opens up new expectations for their use in these diseases. The goal for stem cell-based therapy is to provide a permanent cure. To achieve this, it will be necessary to obtain a cellular product, original or genetically modified, that has a high migration capacity and homes into the intestine, has high survival after transplantation, regulates the immune reaction while not being visible to the patient’s immune system, and repairs the injured tissue.
Collapse
|
99
|
Son S, Liang MS, Lei P, Xue X, Furlani EP, Andreadis ST. Magnetofection Mediated Transient NANOG Overexpression Enhances Proliferation and Myogenic Differentiation of Human Hair Follicle Derived Mesenchymal Stem Cells. Bioconjug Chem 2015; 26:1314-27. [PMID: 25685943 DOI: 10.1021/bc5005203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We used magnetofection (MF) to achieve high transfection efficiency into human mesenchymal stem cells (MSCs). A custom-made magnet array, matching well-to-well to a 24-well plate, was generated and characterized. Theoretical predictions of magnetic force distribution within each well demonstrated that there was no magnetic field interference among magnets in adjacent wells. An optimized protocol for efficient gene delivery to human hair follicle derived MSCs (hHF-MSCs) was established using an egfp-encoding plasmid, reaching approximately ∼50% transfection efficiency without significant cytotoxicity. Then we applied the optimized MF protocol to express the pluripotency-associated transcription factor NANOG, which was previously shown to reverse the effects of organismal aging on MSC proliferation and myogenic differentiation capacity. Indeed, MF-mediated NANOG delivery increased proliferation and enhanced the differentiation of hHF-MSCs into smooth muscle cells (SMCs). Collectively, our results show that MF can achieve high levels of gene delivery to MSCs and, therefore, may be employed to moderate or reverse the effects of cellular senescence or reprogram cells to the pluripotent state without permanent genetic modification.
Collapse
Affiliation(s)
| | | | | | | | | | - Stelios T Andreadis
- ∥Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203, United States
| |
Collapse
|
100
|
Koobatian MT, Liang MS, Swartz DD, Andreadis ST. Differential effects of culture senescence and mechanical stimulation on the proliferation and leiomyogenic differentiation of MSC from different sources: implications for engineering vascular grafts. Tissue Eng Part A 2015; 21:1364-75. [PMID: 25517657 DOI: 10.1089/ten.tea.2014.0535] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We examined the effects of senescence on the proliferation and leiomyogenic differentiation potential of mesenchymal stem cells (MSCs) isolated from bone marrow (BM-MSCs) or hair follicles (HF-MSCs). To this end, we compared ovine HF-MSCs and BM-MSCs in terms of their proliferation and differentiation potential to the smooth muscle cell lineage. We discovered that HF-MSCs are less susceptible to culture senescence compared with BM-MSCs. We hypothesized that application of mechanical forces may enhance the contractility and mechanical properties of vascular constructs prepared from senescent MSCs. Interestingly, HF-MSCs and BM-MSCs responded differently to changes in the mechanical microenvironment, suggesting that despite phenotypic similarities, MSCs from different anatomic locations may activate different pathways in response to the same microenvironmental factors. In turn, this may also suggest that cell-based tissue regeneration approaches may need to be tailored to the stem cell origin, donor age, and culture time for optimal results.
Collapse
Affiliation(s)
- Maxwell T Koobatian
- 1 Department of Physiology and Biophysics, University at Buffalo , The State University of New York, Amherst, New York
| | | | | | | |
Collapse
|