1
|
Farag A, Hendawy H, Emam MH, Hasegawa M, Mandour AS, Tanaka R. Stem Cell Therapies in Canine Cardiology: Comparative Efficacy, Emerging Trends, and Clinical Integration. Biomolecules 2025; 15:371. [PMID: 40149907 PMCID: PMC11940628 DOI: 10.3390/biom15030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular diseases are a leading cause of morbidity and mortality in dogs, with limited options available for reversing myocardial damage. Stem cell therapies have shown significant potential for cardiac repair, owing to their immunomodulatory, antifibrotic, and regenerative properties. This review evaluates the therapeutic applications of mesenchymal stem cells (MSCs) derived from bone marrow, adipose tissue, and Wharton's jelly with a focus on their role in canine cardiology and their immunoregulatory properties. Preclinical studies have highlighted their efficacy in enhancing cardiac function, reducing fibrosis, and promoting angiogenesis. Various delivery methods, including intracoronary and intramyocardial injections, are assessed for their safety and efficacy. Challenges such as low cell retention, differentiation efficiency, and variability in therapeutic responses are also discussed. Emerging strategies, including genetic modifications and combination therapies, aim to enhance the efficacy of MSCs. Additionally, advances in delivery systems and regulatory frameworks are reviewed to support clinical translation. This comprehensive evaluation underscores the potential of stem cell therapies to revolutionize canine cardiovascular disease management while identifying critical areas for future research and clinical integration.
Collapse
Affiliation(s)
- Ahmed Farag
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Hanan Hendawy
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Mahmoud H. Emam
- Animal Medicine Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mizuki Hasegawa
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ryou Tanaka
- Faculty of Agriculture, Veterinary Teaching Hospital, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
2
|
Bonfield TL, Lazarus HM. Human mesenchymal stem cell therapy: Potential advances for reducing cystic fibrosis infection and organ inflammation. Best Pract Res Clin Haematol 2025; 38:101602. [PMID: 40274338 DOI: 10.1016/j.beha.2025.101602] [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: 11/20/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Innovation in cystic fibrosis (CF) supportive care, including implementing new antimicrobial agents, improved physiotherapy, and highly effective modulators therapy, has advanced patient survival into the 4th and 5th decades of life. However, even with these remarkable improvements in therapy, CF patients continue to suffer from pulmonary infection and other visceral organ complications associated with long-term deficient cystic fibrosis transmembrane conductance regulator (CFTR) expression. Human mesenchymal stem cells (MSCs) have been utilized in tissue engineering based upon their capacity to provide structural components of mesenchymal tissues. An alternative role of MSCs, however is their versatile utilization as cell-based infusion powerhouses due to the unique capacity to deliver milieu specific soluble biologic factors, promoting immune supportive antimicrobial and anti-inflammatory potency. MSCs derived from umbilical cord blood, bone marrow, adipose and other tissues can be expanded in ex vivo using good manufacturing procedure facilities for a safe, unique therapeutic to reduce and limit CF infection and facilitate the resolution of multi-organ inflammation. In our efforts, we conducted extensive preclinical development and validation of an allogeneic derived bone marrow derived MSC product in preparation for a clinical trial in CF. In this process, potency models were developed to ensure the functional capacity of the MSC product to provide clinical benefit. In vitro, murine in vivo and patient tissue ex vivo potency models were utilized to follow MSC anti-infective and anti-inflammatory potency associated with the CFTR deficient environment. We showed in our "First in CF" clinical trial that the allogeneic MSCs obtained from healthy volunteer bone marrow samples were safe. The advent of improved CF care measures and exciting new small molecules has changed the survival and morbidity phenotype of patients with CF, however, there are CF patients who cannot tolerate or have genotypes that are non-responsive to modulators. Additionally, even with the small molecule therapy, CF patients are living longer, but without genetic correction, with the CF disease manifestation aggravated by the continuance of pre-existing CFTR-associated clinical issues such as ongoing inflammation. MSCs secrete bio-active factors that enhance and protect tissue function and can promote "self-immune" regulation. These properties can provide therapeutic support for the traditional and changing face of CF disease clinical complications. Further, MSC-derived bio-active factors can directly mitigate colonizing pathogens' survival by producing antimicrobial peptides (AMPs) which change the pathogen surface and increase host recognition, elimination, and sensitivity to antibiotics. Herein, we review the potential of MSC therapeutics for treating many facets of CF, emphasizing the potential for providing great additive therapeutics for managing morbidity and quality of life.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Genetics and Genome Sciences, National Center for Regenerative Medicine, Pediatrics and Pathology, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| | - Hillard M Lazarus
- Department of Medicine, Hematology and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
3
|
Zhang L, Deng Y, Bai X, Wei X, Ren Y, Chen S, Deng H. Cell therapy for end-stage liver disease: Current state and clinical challenge. Chin Med J (Engl) 2024; 137:2808-2820. [PMID: 39602326 DOI: 10.1097/cm9.0000000000003332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT Liver disease involves a complex interplay of pathological processes, including inflammation, hepatocyte necrosis, and fibrosis. End-stage liver disease (ESLD), such as liver failure and decompensated cirrhosis, has a high mortality rate, and liver transplantation is the only effective treatment. However, to overcome problems such as the shortage of donor livers and complications related to immunosuppression, there is an urgent need for new treatment strategies that need to be developed for patients with ESLD. For instance, hepatocytes derived from donor livers or stem cells can be engrafted and multiplied in the liver, substituting the host hepatocytes and rebuilding the liver parenchyma. Stem cell therapy, especially mesenchymal stem cell therapy, has been widely proved to restore liver function and alleviate liver injury in patients with severe liver disease, which has contributed to the clinical application of cell therapy. In this review, we discussed the types of cells used to treat ESLD and their therapeutic mechanisms. We also summarized the progress of clinical trials around the world and provided a perspective on cell therapy.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | |
Collapse
|
4
|
Thamm OC, Eschborn J, Zimmermann L, Dekker C, Martin H, Brockmann M, Zinser MJ, Fuchs PC. Sublesional fat grafting leads to a temporary improvement of wound healing in chronic leg ulcers: A prospective, randomised clinical trial. Wound Repair Regen 2023; 31:663-670. [PMID: 37534628 DOI: 10.1111/wrr.13111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/14/2023] [Accepted: 07/18/2023] [Indexed: 08/04/2023]
Abstract
Chronic wounds remain a therapeutic and financial challenge for physicians and the health care systems. Innovative, inexpensive and effective treatment methods would be of immense value. The sublesional fat grafting could be such treatment, although effectiveness and safety have only been assessed in a few randomised clinical trials. The fat graft was obtained by liposuction, washed with the Coleman method and then injected sublesional and into the wound margins after surgical debridement. For the control group, saline solution was used instead of fat. The primary endpoint was to determine the wound size reduction in both groups. The wounds were measured preoperatively, intraoperatively and 3, 7, 21 and 60 days after the intervention. A p-value of <0.05 was considered significant. Furthermore, histology and microbiology of the wounds and pain were assessed. A temporary effect of the treatment was observed after 14 and 21 days. The wound size reduction was significantly larger in the intervention group, whereas after 60 days, no significant difference was detected between both groups. No adverse events could be reported and the pain level was almost equal in the control and intervention group. Sublesional fat grafting temporarily enhanced healing of chronic wounds. The procedure was safe and the pain level was low. Repeated interventions could lead to complete wound closure, which should be determined in future studies.
Collapse
Affiliation(s)
- Oliver C Thamm
- Clinic for Plastic and Reconstructive Surgery, Helios Hospital Berlin-Buch, Berlin, Germany
- Clinic for Plastic- and Reconstructive Surgery, Hand Surgery, Burn Care Center, University Witten/Herdecke, Cologne-Merheim Medical Center, Cologne, Germany
| | - Johannes Eschborn
- Clinic for Plastic and Reconstructive Surgery, Helios Hospital Berlin-Buch, Berlin, Germany
- Clinic for Plastic- and Reconstructive Surgery, Hand Surgery, Burn Care Center, University Witten/Herdecke, Cologne-Merheim Medical Center, Cologne, Germany
| | - Lucas Zimmermann
- Clinic for Orthopedic and Spinal Surgery, Berit Clinic, Speicher, Switzerland
| | - Clara Dekker
- Clinic for Cardiology, Electrophysiology and Rhytmology, Hospital Porz am Rhein, Cologne, Germany
| | - Hubert Martin
- Institute for Neuropathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Brockmann
- Institute for Pathology, University Witten/Herdecke, Cologne-Merheim Medical Center, Cologne, Germany
| | - Max J Zinser
- Department for Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, University Hospital of Cologne, Cologne, Germany
| | - Paul C Fuchs
- Clinic for Plastic- and Reconstructive Surgery, Hand Surgery, Burn Care Center, University Witten/Herdecke, Cologne-Merheim Medical Center, Cologne, Germany
| |
Collapse
|
5
|
Khan S, Mahgoub S, Fallatah N, Lalor PF, Newsome PN. Liver Disease and Cell Therapy: Advances Made and Remaining Challenges. Stem Cells 2023; 41:739-761. [PMID: 37052348 PMCID: PMC10809282 DOI: 10.1093/stmcls/sxad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 02/27/2023] [Indexed: 04/14/2023]
Abstract
The limited availability of organs for liver transplantation, the ultimate curative treatment for end stage liver disease, has resulted in a growing and unmet need for alternative therapies. Mesenchymal stromal cells (MSCs) with their broad ranging anti-inflammatory and immunomodulatory properties have therefore emerged as a promising therapeutic agent in treating inflammatory liver disease. Significant strides have been made in exploring their biological activity. Clinical application of MSC has shifted the paradigm from using their regenerative potential to one which harnesses their immunomodulatory properties. Reassuringly, MSCs have been extensively investigated for over 30 years with encouraging efficacy and safety data from translational and early phase clinical studies, but questions remain about their utility. Therefore, in this review, we examine the translational and clinical studies using MSCs in various liver diseases and their impact on dampening immune-mediated liver damage. Our key observations include progress made thus far with use of MSCs for clinical use, inconsistency in the literature to allow meaningful comparison between different studies and need for standardized protocols for MSC manufacture and administration. In addition, the emerging role of MSC-derived extracellular vesicles as an alternative to MSC has been reviewed. We have also highlighted some of the remaining clinical challenges that should be addressed before MSC can progress to be considered as therapy for patients with liver disease.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Sara Mahgoub
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Nada Fallatah
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Patricia F Lalor
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Philip N Newsome
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
6
|
Szydlak R. Mesenchymal stem cells in ischemic tissue regeneration. World J Stem Cells 2023; 15:16-30. [PMID: 36909782 PMCID: PMC9993139 DOI: 10.4252/wjsc.v15.i2.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 01/19/2023] [Indexed: 02/21/2023] Open
Abstract
Diseases caused by ischemia are one of the leading causes of death in the world. Current therapies for treating acute myocardial infarction, ischemic stroke, and critical limb ischemia do not complete recovery. Regenerative therapies opens new therapeutic strategy in the treatment of ischemic disorders. Mesenchymal stem cells (MSCs) are the most promising option in the field of cell-based therapies, due to their secretory and immunomodulatory abilities, that contribute to ease inflammation and promote the regeneration of damaged tissues. This review presents the current knowledge of the mechanisms of action of MSCs and their therapeutic effects in the treatment of ischemic diseases, described on the basis of data from in vitro experiments and preclinical animal studies, and also summarize the effects of using these cells in clinical trial settings. Since the obtained therapeutic benefits are not always satisfactory, approaches aimed at enhancing the effect of MSCs in regenerative therapies are presented at the end.
Collapse
Affiliation(s)
- Renata Szydlak
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków 31-034, Poland
| |
Collapse
|
7
|
Bonfield TL, Sutton MT, Fletcher DR, Reese-Koc J, Roesch EA, Lazarus HM, Chmiel JF, Caplan AI. Human Mesenchymal Stem Cell (hMSC) Donor Potency Selection for the "First in Cystic Fibrosis" Phase I Clinical Trial (CEASE-CF). Pharmaceuticals (Basel) 2023; 16:220. [PMID: 37259368 PMCID: PMC9960767 DOI: 10.3390/ph16020220] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 08/27/2023] Open
Abstract
Human Mesenchymal Stem Cell (hMSC) immunotherapy has been shown to provide both anti-inflammatory and anti-microbial effectiveness in a variety of diseases. The clinical potency of hMSCs is based upon an initial direct hMSC effect on the pro-inflammatory and anti-microbial pathophysiology as well as sustained potency through orchestrating the host immunity to optimize the resolution of infection and tissue damage. Cystic fibrosis (CF) patients suffer from a lung disease characterized by excessive inflammation and chronic infection as well as a variety of other systemic anomalies associated with the consequences of abnormal cystic fibrosis transmembrane conductance regulator (CFTR) function. The application of hMSC immunotherapy to the CF clinical armamentarium is important even in the era of modulators when patients with an established disease still need anti-inflammatory and anti-microbial therapies. Additionally, people with CF mutations not addressed by current modulator resources need anti-inflammation and anti-infection management. Furthermore, hMSCs possess dynamic therapeutic properties, but the potency of their products is highly variable with respect to their anti-inflammatory and anti-microbial effects. Due to the variability of hMSC products, we utilized standardized in vitro and in vivo models to select hMSC donor preparations with the greatest potential for clinical efficacy. The models that were used recapitulate many of the pathophysiologic outcomes associated with CF. We applied this strategy in pursuit of identifying the optimal donor to utilize for the "First in CF" Phase I clinical trial of hMSCs as an immunotherapy and anti-microbial therapy for people with cystic fibrosis. The hMSCs screened in this study demonstrated significant diversity in antimicrobial and anti-inflammatory function using models which mimic some aspects of CF infection and inflammation. However, the variability in activity between in vitro potency and in vivo effectiveness continues to be refined. Future studies require and in-depth pursuit of hMSC molecular signatures that ultimately predict the capacity of hMSCs to function in the clinical setting.
Collapse
Affiliation(s)
- Tracey L. Bonfield
- Department of Genetics and Genome Sciences, National Center Regenerative Medicine and Pediatrics, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, BRB 822, Cleveland, OH 444106, USA
- National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 444106, USA
- Department of Pediatric Pulmonary, Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
| | - Morgan T. Sutton
- Department of Genetics and Genome Sciences, National Center Regenerative Medicine and Pediatrics, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, BRB 822, Cleveland, OH 444106, USA
- National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 444106, USA
- Department of Pediatric Pulmonary, Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
- Saint Jude Children’s Research Hospital, Graduate School of Biomedical Sciences, Memphis, TN 38105, USA
| | - David R. Fletcher
- Department of Genetics and Genome Sciences, National Center Regenerative Medicine and Pediatrics, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, BRB 822, Cleveland, OH 444106, USA
- Department of Pediatric Pulmonary, Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
| | - Jane Reese-Koc
- National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 444106, USA
- University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Erica A. Roesch
- Department of Pediatric Pulmonary, Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
| | - Hillard M. Lazarus
- National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 444106, USA
- University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - James F. Chmiel
- Department of Pediatrics, Riley Hospital for Children at IU Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Arnold I. Caplan
- National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 444106, USA
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
8
|
Kankuri E, Karjalainen P, Vento A. Atrial Appendage-Derived Cardiac Micrografts: An Emerging Cellular Therapy for Heart Failure. CARDIOVASCULAR APPLICATIONS OF STEM CELLS 2023:155-181. [DOI: 10.1007/978-981-99-0722-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
9
|
Niebergall-Roth E, Frank NY, Ganss C, Frank MH, Kluth MA. Skin-Derived ABCB5 + Mesenchymal Stem Cells for High-Medical-Need Inflammatory Diseases: From Discovery to Entering Clinical Routine. Int J Mol Sci 2022; 24:66. [PMID: 36613507 PMCID: PMC9820160 DOI: 10.3390/ijms24010066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
The ATP-binding cassette superfamily member ABCB5 identifies a subset of skin-resident mesenchymal stem cells (MSCs) that exhibit potent immunomodulatory and wound healing-promoting capacities along with superior homing ability. The ABCB5+ MSCs can be easily accessed from discarded skin samples, expanded, and delivered as a highly homogenous medicinal product with standardized potency. A range of preclinical studies has suggested therapeutic efficacy of ABCB5+ MSCs in a variety of currently uncurable skin and non-skin inflammatory diseases, which has been substantiated thus far by distinct clinical trials in chronic skin wounds or recessive dystrophic epidermolysis bullosa. Therefore, skin-derived ABCB5+ MSCs have the potential to provide a breakthrough at the forefront of MSC-based therapies striving to fulfill current unmet medical needs. The most recent milestones in this regard are the approval of a phase III pivotal trial of ABCB5+ MSCs for treatment of recessive dystrophic and junctional epidermolysis bullosa by the US Food and Drug Administration, and national market access of ABCB5+ MSCs (AMESANAR®) for therapy-refractory chronic venous ulcers under the national hospital exemption pathway in Germany.
Collapse
Affiliation(s)
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Ganss
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| | - Markus H. Frank
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Mark A. Kluth
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Kerstan A, Dieter K, Niebergall-Roth E, Klingele S, Jünger M, Hasslacher C, Daeschlein G, Stemler L, Meyer-Pannwitt U, Schubert K, Klausmann G, Raab T, Goebeler M, Kraft K, Esterlechner J, Schröder HM, Sadeghi S, Ballikaya S, Gasser M, Waaga-Gasser AM, Murphy GF, Orgill DP, Frank NY, Ganss C, Scharffetter-Kochanek K, Frank MH, Kluth MA. Translational development of ABCB5 + dermal mesenchymal stem cells for therapeutic induction of angiogenesis in non-healing diabetic foot ulcers. Stem Cell Res Ther 2022; 13:455. [PMID: 36064604 PMCID: PMC9444095 DOI: 10.1186/s13287-022-03156-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background While rapid healing of diabetic foot ulcers (DFUs) is highly desirable to avoid infections, amputations and life-threatening complications, DFUs often respond poorly to standard treatment. GMP-manufactured skin-derived ABCB5+ mesenchymal stem cells (MSCs) might provide a new adjunctive DFU treatment, based on their remarkable skin wound homing and engraftment potential, their ability to adaptively respond to inflammatory signals, and their wound healing-promoting efficacy in mouse wound models and human chronic venous ulcers. Methods The angiogenic potential of ABCB5+ MSCs was characterized with respect to angiogenic factor expression at the mRNA and protein level, in vitro endothelial trans-differentiation and tube formation potential, and perfusion-restoring capacity in a mouse hindlimb ischemia model. Finally, the efficacy and safety of ABCB5+ MSCs for topical adjunctive treatment of chronic, standard therapy-refractory, neuropathic plantar DFUs were assessed in an open-label single-arm clinical trial. Results Hypoxic incubation of ABCB5+ MSCs led to posttranslational stabilization of the hypoxia-inducible transcription factor 1α (HIF-1α) and upregulation of HIF-1α mRNA levels. HIF-1α pathway activation was accompanied by upregulation of vascular endothelial growth factor (VEGF) transcription and increase in VEGF protein secretion. Upon culture in growth factor-supplemented medium, ABCB5+ MSCs expressed the endothelial-lineage marker CD31, and after seeding on gel matrix, ABCB5+ MSCs demonstrated formation of capillary-like structures comparable with human umbilical vein endothelial cells. Intramuscularly injected ABCB5+ MSCs to mice with surgically induced hindlimb ischemia accelerated perfusion recovery as measured by laser Doppler blood perfusion imaging and enhanced capillary proliferation and vascularization in the ischemic muscles. Adjunctive topical application of ABCB5+ MSCs onto therapy-refractory DFUs elicited median wound surface area reductions from baseline of 59% (full analysis set, n = 23), 64% (per-protocol set, n = 20) and 67% (subgroup of responders, n = 17) at week 12, while no treatment-related adverse events were observed. Conclusions The present observations identify GMP-manufactured ABCB5+ dermal MSCs as a potential, safe candidate for adjunctive therapy of otherwise incurable DFUs and justify the conduct of a larger, randomized controlled trial to validate the clinical efficacy. Trial registration: ClinicalTrials.gov, NCT03267784, Registered 30 August 2017, https://clinicaltrials.gov/ct2/show/NCT03267784 Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03156-9.
Collapse
Affiliation(s)
- Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Sabrina Klingele
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Michael Jünger
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany
| | | | - Georg Daeschlein
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany.,Clinic of Dermatology, Immunology and Allergology, Medical University Brandenburg "Theodor Fontane" Medical Center Dessau, Dessau, Germany
| | - Lutz Stemler
- Diabetologikum DDG Ludwigshafen, Ludwigshafen, Germany
| | | | | | | | | | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Samar Sadeghi
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Seda Ballikaya
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Martin Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Ana M Waaga-Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany.,Division of Renal (Kidney) Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George F Murphy
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Natasha Y Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA.,Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Christoph Ganss
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Mark A Kluth
- RHEACELL GmbH & Co. KG, Heidelberg, Germany. .,TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.
| |
Collapse
|
11
|
Boada-Pladellorens A, Avellanet M, Pages-Bolibar E, Veiga A. Stromal vascular fraction therapy for knee osteoarthritis: a systematic review. Ther Adv Musculoskelet Dis 2022; 14:1759720X221117879. [PMID: 35991523 PMCID: PMC9386815 DOI: 10.1177/1759720x221117879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/19/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Regenerative cell therapies, such as adipose-derived stromal vascular fraction (SVF), have been postulated as potential treatments for knee osteoarthritis (KOA). Objectives: To assess the efficacy and safety of SVF treatment against placebo and other standard therapies for treating KOA in adult patients. Design: A systematic review. Data sources and methods: We searched the following databases: MEDLINE via PubMed, Epistemonikos, PEDro, DynaMed, TripDatabase, Elsevier via Clinicalkey and Cochrane Controlled Trials Register. We included prospective interventional studies where treatment with SVF in adults with KOA was compared against placebo or other standard therapies, and results were objectively measured with at least one widely recognised osteoarthritis scale. Results: Among 266 studies published until May 2021, nine met our inclusion criteria. A total of 239 patients (274 knees) were included in our study. The follow-up ranged from 6 to 24 months. Six studies had a control group (only one being placebo). All studies showed that SVF improved pain and functionality measured, in most cases, with the visual analogue scale and the Western Ontario and McMaster Universities Osteoarthritis Index. In addition, five studies reported an improvement in anatomical structures, as detected in MR images. However, the number of cells contained in SVF varied substantially between different studies, which could induce a comparison bias. Conclusion: Although based on a small number of dissimilar studies, SVF was considered a safe treatment for KOA and could be promising in terms of pain, functionality and anatomical structure improvement. However, SVF products need to be standardised, the number of cells homogenised and the use of concomitant treatments reduced to establish proper comparisons. Registration: PROSPERO registration number: CRD42021284187.
Collapse
Affiliation(s)
- Anna Boada-Pladellorens
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Carrer dels Escalls, AD700 Escaldes-Engordany, Andorra
| | - Mercè Avellanet
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Escaldes-Engordany, Andorra
| | - Esther Pages-Bolibar
- Physical Medicine and Rehabilitation Department, Hospital Nostra Senyora de Meritxell, Escaldes-Engordany, Andorra
| | - Anna Veiga
- Barcelona Stem Cell Bank, Regenerative Medicine Programme, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, Barcelona, Spain
| |
Collapse
|
12
|
Kerstan A, Dieter K, Niebergall-Roth E, Dachtler AK, Kraft K, Stücker M, Daeschlein G, Jünger M, Görge T, Meyer-Pannwitt U, Erfurt-Berge C, von Engelhardt C, Klare A, Pfeiffer C, Esterlechner J, Schröder HM, Gasser M, Waaga-Gasser AM, Goebeler M, Ballikaya S, Sadeghi S, Murphy GF, Orgill DP, Frank NY, Ganss C, Scharffetter-Kochanek K, Frank MH, Kluth MA. Allogeneic ABCB5 + mesenchymal stem cells for treatment-refractory chronic venous ulcers: a phase I/IIa clinical trial. JID INNOVATIONS 2022; 2:100067. [PMID: 34870260 PMCID: PMC8635035 DOI: 10.1016/j.xjidi.2021.100067] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
A significant number of chronic venous ulcers (CVUs) fail to heal despite of guideline-conform standard of care. Skin-derived ABCB5+ mesenchymal stem cells (MSCs) can dampen the sustained IL-1β-driven inflammation present in chronic wounds. Based on their wound healing-facilitating effects in a mouse CVU model and an autologous first-in-human study, ABCB5+ MSCs have emerged as a potential candidate for cell-based advanced therapy of non-healing CVUs. In the present interventional, multicenter, single-arm, phase I/IIa clinical trial, subjects whose CVU had emerged as standard therapy-resistant received one or two topical applications of 1×106 allogeneic ABCB5+ MSCs/cm2 wound area in addition to standard treatment. Out of 83 treatment-emergent adverse events, only three were judged related to the cell product; they were mild or moderate and recovered without sequelae. Wound size markedly decreased from baseline to week 12, resulting in a median wound size reduction of 76% (full analysis set, N=31), 78% (per-protocol set, N=27) and 87% (subset of responders; n=21). In conclusion, the study treatment was well tolerated and safe. The treatment elicited a profound wound size reduction within 12 weeks, identifying ABCB5+ MSCs as a potential candidate for adjunctive therapy of otherwise incurable CVUs. These results justify the conduct of a larger, randomized, controlled trial to confirm clinical efficacy.
Collapse
Affiliation(s)
- Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | | | - Markus Stücker
- Department of Dermatology, St. Josef Hospital, Catholic Clinic Bochum, Ruhr University Bochum, Bochum, Germany
| | - Georg Daeschlein
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany
- Clinic of Dermatology, Immunology and Allergology, Medical University Brandenburg “Theodor Fontane” Medical Center Dessau, Dessau, Germany
| | - Michael Jünger
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany
| | - Tobias Görge
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Ulrich Meyer-Pannwitt
- pro scientia med at the Department of Clinical Research and Development, MARE Clinic, Kiel, Germany
| | | | | | | | - Christiane Pfeiffer
- Department of Dermatology and Allergic Diseases, University Hospital, Ulm, Germany
| | | | | | - Martin Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Ana M. Waaga-Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany
- Division of Renal (Kidney) Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | - George F. Murphy
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dennis P. Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | | | | | - Markus H. Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Mark A. Kluth
- RHEACELL, Heidelberg, Germany
- TICEBA, Heidelberg, Germany
| |
Collapse
|
13
|
González-González A, García-Sánchez D, Alfonso-Fernández A, Haider KH, Rodríguez-Rey JC, Pérez-Campo FM. Regenerative Medicine Applied to the Treatment of Musculoskeletal Pathologies. HANDBOOK OF STEM CELL THERAPY 2022:1123-1158. [DOI: 10.1007/978-981-19-2655-6_50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Caplan AI. Mesenchymal stem cells and COVID-19: the process of discovery and of translation. BIOMATERIALS TRANSLATIONAL 2021; 2:307-311. [PMID: 35837414 PMCID: PMC9255802 DOI: 10.12336/biomatertransl.2021.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/19/2021] [Accepted: 11/16/2021] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells were developed as a cell-based therapeutic in the 1990's. The translation of culture expanded mesenchymal stem cells from a basic science focus into a modern therapeutic has taken 30 years. The current state of the basic science information argues that mesenchymal stem cells may be curative for coronavirus disease 2019 (COVID-19). Indeed, early small-scale clinical trials have shown positive results. The issue raised is how to assemble the resources to get this cell-based therapy approved for clinical use. The technology is complex, the COVID-19 viral infections are life threatening, the cost is high, but human life is precious. What will it take to perfect this potentially curative technology?
Collapse
|
15
|
Kiritsi D, Dieter K, Niebergall-Roth E, Fluhr S, Daniele C, Esterlechner J, Sadeghi S, Ballikaya S, Erdinger L, Schauer F, Gewert S, Laimer M, Bauer JW, Hovnanian A, Zambruno G, El Hachem M, Bourrat E, Papanikolaou M, Petrof G, Kitzmüller S, Ebens CL, Frank MH, Frank NY, Ganss C, Martinez AE, McGrath JA, Tolar J, Kluth MA. Clinical trial of ABCB5+ mesenchymal stem cells for recessive dystrophic epidermolysis bullosa. JCI Insight 2021; 6:151922. [PMID: 34665781 PMCID: PMC8663784 DOI: 10.1172/jci.insight.151922] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/13/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB) is a rare, devastating, and life-threatening inherited skin fragility disorder that comes about due to a lack of functional type VII collagen, for which no effective therapy exists. ABCB5+ dermal mesenchymal stem cells (ABCB5+ MSCs) possess immunomodulatory, inflammation-dampening, and tissue-healing capacities. In a Col7a1–/– mouse model of RDEB, treatment with ABCB5+ MSCs markedly extended the animals’ lifespans. METHODS In this international, multicentric, single-arm, phase I/IIa clinical trial, 16 patients (aged 4–36 years) enrolled into 4 age cohorts received 3 i.v. infusions of 2 × 106 ABCB5+ MSCs/kg on days 0, 17, and 35. Patients were followed up for 12 weeks regarding efficacy and 12 months regarding safety. RESULTS At 12 weeks, statistically significant median (IQR) reductions in the Epidermolysis Bullosa Disease Activity and Scarring Index activity (EBDASI activity) score of 13.0% (2.9%–30%; P = 0.049) and the Instrument for Scoring Clinical Outcome of Research for Epidermolysis Bullosa clinician (iscorEB‑c) score of 18.2% (1.9%–39.8%; P = 0.037) were observed. Reductions in itch and pain numerical rating scale scores were greatest on day 35, amounting to 37.5% (0.0%–42.9%; P = 0.033) and 25.0% (–8.4% to 46.4%; P = 0.168), respectively. Three adverse events were considered related to the cell product: 1 mild lymphadenopathy and 2 hypersensitivity reactions. The latter 2 were serious but resolved without sequelae shortly after withdrawal of treatment. CONCLUSION This trial demonstrates good tolerability, manageable safety, and potential efficacy of i.v. ABCB5+ MSCs as a readily available disease-modifying therapy for RDEB and provides a rationale for further clinical evaluation. TRIAL REGISTRATION Clinicaltrials.gov NCT03529877; EudraCT 2018-001009-98. FUNDING The trial was sponsored by RHEACELL GmbH & Co. KG. Contributions by NYF and MHF to this work were supported by the NIH/National Eye Institute (NEI) grants RO1EY025794 and R24EY028767.
Collapse
Affiliation(s)
- Dimitra Kiritsi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | | | | | | | | | | | | | | | | | - Franziska Schauer
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Stella Gewert
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Martin Laimer
- EB House Austria, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Johann W Bauer
- EB House Austria, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Alain Hovnanian
- Department of Genetics at Necker Hospital and.,Department of Dermatology at Saint-Louis Hospital, INSERM UMR
| | | | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Emmanuelle Bourrat
- Department of Dermatology, Reference Center for Rare Skin Diseases MAGEC, St. Louis Hospital, Paris, France
| | - Maria Papanikolaou
- St. John's Institute of Dermatology, Guy's Hospital, King's College London, London, United Kingdom
| | - Gabriela Petrof
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Sophie Kitzmüller
- EB House Austria, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Christen L Ebens
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA.,School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Natasha Y Frank
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA.,Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.,Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christoph Ganss
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Heidelberg, Germany
| | - Anna E Martinez
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - John A McGrath
- St. John's Institute of Dermatology, Guy's Hospital, King's College London, London, United Kingdom
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Mark A Kluth
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Heidelberg, Germany
| |
Collapse
|
16
|
van Heeckeren AM, Sutton MT, Fletcher DR, Hodges CA, Caplan AI, Bonfield TL. Enhancing Cystic Fibrosis Immune Regulation. Front Pharmacol 2021; 12:573065. [PMID: 34054509 PMCID: PMC8155373 DOI: 10.3389/fphar.2021.573065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
In cystic fibrosis (CF), sustained infection and exuberant inflammation results in debilitating and often fatal lung disease. Advancement in CF therapeutics has provided successful treatment regimens for a variety of clinical consequences in CF; however effective means to treat the pulmonary infection and inflammation continues to be problematic. Even with the successful development of small molecule cystic fibrosis transmembrane conductance regulator (CFTR) correctors and potentiators, there is only a modest effect on established infection and inflammation in CF patients. In the pursuit of therapeutics to treat inflammation, the conundrum to address is how to overcome the inflammatory response without jeopardizing the required immunity to manage pathogens and prevent infection. The key therapeutic would have the capacity to dull the inflammatory response, while sustaining the ability to manage infections. Advances in cell-based therapy have opened up the avenue for dynamic and versatile immune interventions that may support this requirement. Cell based therapy has the capacity to augment the patient’s own ability to manage their inflammatory status while at the same time sustaining anti-pathogen immunity. The studies highlighted in this manuscript outline the potential use of cell-based therapy for CF. The data demonstrate that 1) total bone marrow aspirates containing Cftr sufficient hematopoietic and mesenchymal stem cells (hMSCs) provide Cftr deficient mice >50% improvement in survival and improved management of infection and inflammation; 2) myeloid cells can provide sufficient Cftr to provide pre-clinical anti-inflammatory and antimicrobial benefit; 3) hMSCs provide significant improvement in survival and management of infection and inflammation in CF; 4) the combined interaction between macrophages and hMSCs can potentially enhance anti-inflammatory and antimicrobial support through manipulating PPARγ. These data support the development of optimized cell-based therapeutics to enhance CF patient’s own immune repertoire and capacity to maintain the balance between inflammation and pathogen management.
Collapse
Affiliation(s)
- Anna M van Heeckeren
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Morgan T Sutton
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, TN, United States
| | - David R Fletcher
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Craig A Hodges
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Arnold I Caplan
- Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tracey L Bonfield
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
17
|
Bonfield TL, Sutton MT, Fletcher DR, Folz MA, Ragavapuram V, Somoza RA, Caplan AI. Donor-defined mesenchymal stem cell antimicrobial potency against nontuberculous mycobacterium. Stem Cells Transl Med 2021; 10:1202-1216. [PMID: 33943038 PMCID: PMC8284776 DOI: 10.1002/sctm.20-0521] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic nontuberculous mycobacterial infections with Mycobacterium avium and Mycobacterium intracellulare complicate bronchiectasis, chronic obstructive airway disease, and the health of aging individuals. These insidious intracellular pathogens cause considerable morbidity and eventual mortality in individuals colonized with these bacteria. Current treatment regimens with antibiotic macrolides are both toxic and often inefficient at providing infection resolution. In this article, we demonstrate that human marrow‐derived mesenchymal stem cells are antimicrobial and anti‐inflammatory in vitro and in the context of an in vivo sustained infection of either M. avium and/or M. intracellulare.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Morgan T Sutton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA.,St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, Tennessee, USA
| | - David R Fletcher
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael A Folz
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vaishnavi Ragavapuram
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
18
|
Caplan AI. Placebo Controls: Now??? Arch Immunol Ther Exp (Warsz) 2021; 69:9. [PMID: 33782781 PMCID: PMC8006883 DOI: 10.1007/s00005-021-00612-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/22/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Arnold I Caplan
- Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
19
|
Lohmann S, Eijken M, Møldrup U, Møller BK, Hunter J, Moers C, Leuvenink H, Ploeg RJ, Clahsen-van Groningen MC, Hoogduijn M, Baan CC, Keller AK, Jespersen B. Ex Vivo Administration of Mesenchymal Stromal Cells in Kidney Grafts Against Ischemia-reperfusion Injury-Effective Delivery Without Kidney Function Improvement Posttransplant. Transplantation 2021; 105:517-528. [PMID: 32956281 DOI: 10.1097/tp.0000000000003429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mesenchymal stromal cell (MSC) therapy may improve renal function after ischemia-reperfusion injury in transplantation. Ex vivo renal intraarterial administration is a targeted delivery method, avoiding the lung vasculature, a known barrier for cellular therapies. In a randomized and blinded study, we tested the feasibility and effectiveness of MSC therapy in a donation after circulatory death autotransplantation model to improve posttransplant kidney function, using an ex vivo MSC delivery method similar to the clinical standard procedure of pretransplant cold graft flush. METHODS Kidneys exposed to 75 minutes of warm ischemia and 16 hours of static cold storage were intraarterially infused ex vivo with 10 million male porcine MSCs (Tx-MSC, n = 8) or vehicle (Tx-control, n = 8). Afterwards, the kidneys were autotransplanted after contralateral nephrectomy. Biopsies an hour after reperfusion confirmed the presence of MSCs in the renal cortex. Animals were observed for 14 days. RESULTS Postoperatively, peak plasma creatinine was 1230 and 1274 µmol/L (Tx-controls versus Tx-MSC, P = 0.69). During follow-up, no significant differences over time were detected between groups regarding plasma creatinine, plasma neutrophil gelatinase-associated lipocalin, or urine neutrophil gelatinase-associated lipocalin/creatinine ratio. At day 14, measured glomerular filtration rates were 40 and 44 mL/min, P = 0.66. Renal collagen content and fibrosis-related mRNA expression were increased in both groups but without significant differences between the groups. CONCLUSIONS We demonstrated intraarterial MSC infusion to transplant kidneys as a safe and effective method to deliver MSCs to the graft. However, we could not detect any positive effects of this cell treatment within 14 days of observation.
Collapse
Affiliation(s)
- Stine Lohmann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Ulla Møldrup
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - James Hunter
- Nuffield Department of Surgical Sciences, Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Cyril Moers
- Department of Surgery-Organ Donation and Transplantation, University of Medical Center Groningen, Groningen, the Netherlands
| | - Henri Leuvenink
- Department of Surgery-Organ Donation and Transplantation, University of Medical Center Groningen, Groningen, the Netherlands
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences, Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Martin Hoogduijn
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Anna Krarup Keller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bente Jespersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
20
|
González-González A, García-Sánchez D, Dotta M, Rodríguez-Rey JC, Pérez-Campo FM. Mesenchymal stem cells secretome: The cornerstone of cell-free regenerative medicine. World J Stem Cells 2020; 12:1529-1552. [PMID: 33505599 PMCID: PMC7789121 DOI: 10.4252/wjsc.v12.i12.1529] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/07/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the most frequently used stem cells in clinical trials due to their easy isolation from various adult tissues, their ability of homing to injury sites and their potential to differentiate into multiple cell types. However, the realization that the beneficial effect of MSCs relies mainly on their paracrine action, rather than on their engraftment in the recipient tissue and subsequent differentiation, has opened the way to cell-free therapeutic strategies in regenerative medicine. All the soluble factors and vesicles secreted by MSCs are commonly known as secretome. MSCs secretome has a key role in cell-to-cell communication and has been proven to be an active mediator of immune-modulation and regeneration both in vitro and in vivo. Moreover, the use of secretome has key advantages over cell-based therapies, such as a lower immunogenicity and easy production, handling and storage. Importantly, MSCs can be modulated to alter their secretome composition to better suit specific therapeutic goals, thus, opening a large number of possibilities. Altogether these advantages now place MSCs secretome at the center of an important number of investigations in different clinical contexts, enabling rapid scientific progress in this field.
Collapse
Affiliation(s)
- Alberto González-González
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Daniel García-Sánchez
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Monica Dotta
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - José C Rodríguez-Rey
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Flor M Pérez-Campo
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain.
| |
Collapse
|
21
|
Byrnes D, Masterson CH, Artigas A, Laffey JG. Mesenchymal Stem/Stromal Cells Therapy for Sepsis and Acute Respiratory Distress Syndrome. Semin Respir Crit Care Med 2020; 42:20-39. [PMID: 32767301 DOI: 10.1055/s-0040-1713422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sepsis and acute respiratory distress syndrome (ARDS) constitute devastating conditions with high morbidity and mortality. Sepsis results from abnormal host immune response, with evidence for both pro- and anti-inflammatory activation present from the earliest phases. The "proinflammatory" response predominates initially causing host injury, with later-phase sepsis characterized by immune cell hypofunction and opportunistic superinfection. ARDS is characterized by inflammation and disruption of the alveolar-capillary membrane leading to injury and lung dysfunction. Sepsis is the most common cause of ARDS. Approximately 20% of deaths worldwide in 2017 were due to sepsis, while ARDS occurs in over 10% of all intensive care unit patients and results in a mortality of 30 to 45%. Given the fact that sepsis and ARDS share some-but not all-underlying pathophysiologic injury mechanisms, the lack of specific therapies, and their frequent coexistence in the critically ill, it makes sense to consider therapies for both conditions together. In this article, we will focus on the therapeutic potential of mesenchymal stem/stromal cells (MSCs). MSCs are available from several tissues, including bone marrow, umbilical cord, and adipose tissue. Allogeneic administration is feasible, an important advantage for acute conditions like sepsis or ARDS. They possess diverse mechanisms of action of relevance to sepsis and ARDS, including direct and indirect antibacterial actions, potent effects on the innate and adaptive response, and pro-reparative effects. MSCs can be preactivated thereby potentiating their effects, while the use of their extracellular vesicles can avoid whole cell administration. While early-phase clinical trials suggest safety, considerable challenges exist in moving forward to phase III efficacy studies, and to implementation as a therapy should they prove effective.
Collapse
Affiliation(s)
- Declan Byrnes
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Claire H Masterson
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Antonio Artigas
- Critical Care Center, Corporació Sanitaria Parc Tauli, CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | - John G Laffey
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,Department of Anaesthesia, SAOLTA University Health Group, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
22
|
Rai MF, Cheverud JM, Schmidt EJ, Sandell LJ. Genetic correlations between cartilage regeneration and degeneration reveal an inverse relationship. Osteoarthritis Cartilage 2020; 28:1111-1120. [PMID: 32437968 PMCID: PMC7387169 DOI: 10.1016/j.joca.2020.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The etiology of osteoarthritis (OA) is unknown, however, there appears to be a significant contribution from genetics. We have identified recombinant inbred strains of mice derived from LG/J (large) and SM/J (small) strains that vary significantly in their ability to repair articular cartilage and susceptibility to post-traumatic OA due to their genetic composition. Here, we report cartilage repair phenotypes in the same strains of mice in which OA susceptibility was analyzed previously, and determine the genetic correlations between phenotypes. DESIGN We used 12 recombinant inbred strains, including the parental strains, to test three phenotypes: ear-wound healing (n = 263), knee articular cartilage repair (n = 131), and post-traumatic OA (n = 53) induced by the surgical destabilization of the medial meniscus (DMM). Genetic correlations between various traits were calculated as Pearson's correlation coefficients of strain means. RESULTS We found a significant positive correlation between ear-wound healing and articular cartilage regeneration (r = 0.71; P = 0.005). We observed a strong inverse correlation between articular cartilage regeneration and susceptibility to OA based on maximum (r = -0.54; P = 0.036) and summed Osteoarthritis Research Society International (OARSI) scores (r = -0.56; P = 0.028). Synovitis was not significantly correlated with articular cartilage regeneration but was significantly positively correlated with maximum (r = 0.63; P = 0.014) and summed (r = 0.70; P = 0.005) OARSI scores. Ectopic calcification was significantly positively correlated with articular cartilage regeneration (r = 0.59; P = 0.021). CONCLUSIONS Using recombinant inbred strains, our study allows, for the first time, the measurement of genetic correlations of regeneration phenotypes with degeneration phenotypes, characteristic of OA (cartilage degeneration, synovitis). We demonstrate that OA is positively correlated with synovitis and inversely correlated with the ability to repair cartilage. These results suggest an addition to the risk paradigm for OA from a focus on degeneration to regeneration.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Cheverud
- Department of Biology, Loyola University, Chicago, IL, United States
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg, Lynchburg, VA, United States
| | - Linda J. Sandell
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
23
|
Disease-modifying therapies in amyotrophic lateral sclerosis. Neuropharmacology 2020; 167:107986. [DOI: 10.1016/j.neuropharm.2020.107986] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 02/08/2023]
|
24
|
Gomez-Salazar M, Gonzalez-Galofre ZN, Casamitjana J, Crisan M, James AW, Péault B. Five Decades Later, Are Mesenchymal Stem Cells Still Relevant? Front Bioeng Biotechnol 2020; 8:148. [PMID: 32185170 PMCID: PMC7058632 DOI: 10.3389/fbioe.2020.00148] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mario Gomez-Salazar
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zaniah N Gonzalez-Galofre
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Joan Casamitjana
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mihaela Crisan
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Bruno Péault
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
25
|
Autologous Microfragmented Adipose Tissue Reduces the Catabolic and Fibrosis Response in an In Vitro Model of Tendon Cell Inflammation. Stem Cells Int 2019; 2019:5620286. [PMID: 31885616 PMCID: PMC6915130 DOI: 10.1155/2019/5620286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/17/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) emerged as a promising therapy for tendon pathologies. Microfragmented adipose tissue (μFAT) represents a convenient autologous product for the application of MSC-based therapies in the clinical setting. In the present study, the ability of μFAT to counteract inflammatory processes induced by IL-1β on human tendon cells (TCs) was evaluated. Methods Cell viability and proliferation were evaluated after 48 hours of transwell coculture of TCs and autologous μFAT in the presence or absence of IL-1β. Gene expression of scleraxis, collagen type I and type III, metalloproteinases-1 and -3, and cyclooxygenase-2 was evaluated by real-time RT-PCR. The content of VEGF, IL-1Ra, TNFα, and IL-6 was evaluated by ELISA. Results IL-1β-treated TCs showed augmented collagen type III, metalloproteases, and cyclooxygenase-2 expression. μFAT was able to reduce the expression of collagen type III and metalloproteases-1 in a significant manner, and at the same time, it enhanced the production of VEGF, IL-1Ra, and IL-6. Conclusions In this in vitro model of tendon cell inflammation, the paracrine action of μFAT, exerted by anti-inflammatory molecules and growth factors, was able to inhibit the expression of fibrosis and catabolic markers. Then, these results suggest that the application of μFAT may represent an effective conservative or adjuvant therapy for the treatment of tendon disorders.
Collapse
|
26
|
Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019; 4:22. [PMID: 31815001 PMCID: PMC6889290 DOI: 10.1038/s41536-019-0083-6] [Citation(s) in RCA: 1206] [Impact Index Per Article: 201.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.
Collapse
|