1
|
Hoseinzadeh A, Esmaeili SA, Sahebi R, Melak AM, Mahmoudi M, Hasannia M, Baharlou R. Fate and long-lasting therapeutic effects of mesenchymal stromal/stem-like cells: mechanistic insights. Stem Cell Res Ther 2025; 16:33. [PMID: 39901306 PMCID: PMC11792531 DOI: 10.1186/s13287-025-04158-z] [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: 04/27/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
A large body of evidence suggests that mesenchymal stromal cells (MSCs) are able to respond rapidly to the cytokine milieu following systemic infusion. This encounter has the potential to dictate their therapeutic efficacy (also referred to as licensing). MSCs are able to rapidly react to cellular damage by migrating to the inflamed tissue and ultimately modifying the inflammatory microenvironment. However, the limited use of MSCs in clinical practice can be attributed to a lack of understanding of the fate of MSCs in patients after administration and long term MSC-derived therapeutic activity. While the known physiological effectors of viable MSCs make a relative contribution, an innate property of MSCs as a therapeutic agent is their caspase-dependent cell death. These mechanisms may be involving the functional reprogramming of myeloid phagocytes via efferocytosis, the process by which apoptotic bodies (ABs) are identified for engulfment by both specialized and non-specialized phagocytic cells. Recent studies have provided evidence that the uptake of ABs with a distinct genetic component can induce changes in gene expression through the process of epigenetic remodeling. This phenomenon, known as 'trained immunity', has a significant impact on immunometabolism processes. It is hypothesized that the diversity of recipient cells within the inflammatory stroma adjacent to MSCs may potentially serve as a biomarker for predicting the clinical outcome of MSC treatment, while also contributing to the variable outcomes observed with MSC-based therapies. Therefore, the long-term reconstructive process of MSCs may potentially be mediated by MSC apoptosis and subsequent phagocyte-mediated efferocytosis.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed-Alireza Esmaeili
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Sahebi
- Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahmoud Mahmoudi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
2
|
Trapana J, Weinerman J, Lee D, Sedani A, Constantinescu D, Best TM, Hornicek FJ, Hare JM. Cell-based therapy in the treatment of musculoskeletal diseases. Stem Cells Transl Med 2024; 13:959-978. [PMID: 39226104 PMCID: PMC11465182 DOI: 10.1093/stcltm/szae049] [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: 03/13/2023] [Accepted: 05/21/2024] [Indexed: 09/04/2024] Open
Abstract
A limited number of tissues can spontaneously regenerate following injury, and even fewer can regenerate to a state comparable to mature, healthy adult tissue. Mesenchymal stem cells (MSCs) were first described in the 1960s-1970s by Friedenstein et al as a small population of bone marrow cells with osteogenic potential and abilities to differentiate into chondrocytes. In 1991, Arnold Caplan coined the term "mesenchymal cells" after identifying these cells as a theoretical precursor to bone, cartilage, tendon, ligament, marrow stroma, adipocyte, dermis, muscle, and connective tissues. MSCs are derived from periosteum, fat, and muscle. Another attractive property of MSCs is their immunoregulatory and regenerative properties, which result from crosstalk with their microenvironment and components of the innate immune system. Collectively, these properties make MSCs potentially attractive for various therapeutic purposes. MSCs offer potential in sports medicine, aiding in muscle recovery, meniscal tears, and tendon and ligament injuries. In joint disease, MSCs have the potential for chondrogenesis and reversing the effects of osteoarthritis. MSCs have also demonstrated potential application to the treatment of degenerative disc disease of the cervical, thoracic, and lumbar spine.
Collapse
Affiliation(s)
- Justin Trapana
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Jonathan Weinerman
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Danny Lee
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Anil Sedani
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - David Constantinescu
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Thomas M Best
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Francis J Hornicek
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
3
|
Yuan Y, Liu T. Influence of mesenchymal stem cells from different origins on the therapeutic effectiveness of systemic lupus erythematosus. Exp Cell Res 2024; 442:114263. [PMID: 39307406 DOI: 10.1016/j.yexcr.2024.114263] [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: 06/17/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is a chronic autoimmune inflammatory disorder characterized by alterations in the balance between inflammatory and regulatory cytokines. Mesenchymal stem cells (MSCs), which are non-hematopoietic stem cells with multipotent differentiation potential, due to their immunomodulatory, tissue repair, low immunogenicity, and chemotactic properties, have garnered increasing interest in SLE treatment. Studies increasingly reveal the heterogeneous nature of MSC populations. With sources including dental pulp, adipose tissue, bone marrow, and umbilical cord, the therapeutic effects of MSCs on SLE vary depending on their origin. This review consolidates clinical research on MSCs from different sources in treating SLE and analyzes the possible causes underlying these variable outcomes. Additionally, it elucidates five potential factors impacting the outcomes of MSC therapy in SLE: the influence of the microenvironment on MSCs, the complexity and paradoxical aspects of MSC mechanisms in SLE treatment, the heterogeneity of MSCs, the in vivo differentiation potential and post-transplant survival rates of MSCs, and disparities in MSC preparation conditions.
Collapse
Affiliation(s)
- Yuan Yuan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China.
| | - Tong Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China
| |
Collapse
|
4
|
Shan Y, Zhang M, Tao E, Wang J, Wei N, Lu Y, Liu Q, Hao K, Zhou F, Wang G. Pharmacokinetic characteristics of mesenchymal stem cells in translational challenges. Signal Transduct Target Ther 2024; 9:242. [PMID: 39271680 PMCID: PMC11399464 DOI: 10.1038/s41392-024-01936-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024] Open
Abstract
Over the past two decades, mesenchymal stem/stromal cell (MSC) therapy has made substantial strides, transitioning from experimental clinical applications to commercial products. MSC therapies hold considerable promise for treating refractory and critical conditions such as acute graft-versus-host disease, amyotrophic lateral sclerosis, and acute respiratory distress syndrome. Despite recent successes in clinical and commercial applications, MSC therapy still faces challenges when used as a commercial product. Current detection methods have limitations, leaving the dynamic biodistribution, persistence in injured tissues, and ultimate fate of MSCs in patients unclear. Clarifying the relationship between the pharmacokinetic characteristics of MSCs and their therapeutic effects is crucial for patient stratification and the formulation of precise therapeutic regimens. Moreover, the development of advanced imaging and tracking technologies is essential to address these clinical challenges. This review provides a comprehensive analysis of the kinetic properties, key regulatory molecules, different fates, and detection methods relevant to MSCs and discusses concerns in evaluating MSC druggability from the perspective of integrating pharmacokinetics and efficacy. A better understanding of these challenges could improve MSC clinical efficacy and speed up the introduction of MSC therapy products to the market.
Collapse
Affiliation(s)
- Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Mengying Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Enxiang Tao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Yi Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Liu
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
5
|
Pharoun J, Berro J, Sobh J, Abou-Younes MM, Nasr L, Majed A, Khalil A, Joseph, Stephan, Faour WH. Mesenchymal stem cells biological and biotechnological advances: Implications for clinical applications. Eur J Pharmacol 2024; 977:176719. [PMID: 38849038 DOI: 10.1016/j.ejphar.2024.176719] [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: 01/30/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into multiple lineages including bone, cartilage, muscle and fat. They hold immunomodulatory properties and therapeutic ability to treat multiple diseases, including autoimmune and chronic degenerative diseases. In this article, we reviewed the different biological properties, applications and clinical trials of MSCs. Also, we discussed the basics of manufacturing conditions, quality control, and challenges facing MSCs in the clinical setting. METHODS Extensive review of the literature was conducted through the databases PubMed, Google Scholar, and Cochrane. Papers published since 2015 and covering the clinical applications and research of MSC therapy were considered. Furthermore, older papers were considered when referring to pioneering studies in the field. RESULTS The most widely studied stem cells in cell therapy and tissue repair are bone marrow-derived mesenchymal stem cells. Adipose tissue-derived stem cells became more common and to a lesser extent other stem cell sources e.g., foreskin derived MSCs. MSCs therapy were also studied in the setting of COVID-19 infections, ischemic strokes, autoimmune diseases, tumor development and graft rejection. Multiple obstacles, still face the standardization and optimization of MSC therapy such as the survival and the immunophenotype and the efficiency of transplanted cells. MSCs used in clinical settings displayed heterogeneity in their function despite their extraction from healthy donors and expression of similar surface markers. CONCLUSION Mesenchymal stem cells offer a rising therapeutic promise in various diseases. However, their potential use in clinical applications requires further investigation.
Collapse
Affiliation(s)
- Jana Pharoun
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jana Berro
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jeanine Sobh
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | | | - Leah Nasr
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Ali Majed
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Alia Khalil
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Joseph
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Stephan
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36.
| |
Collapse
|
6
|
Eldaly AS, Avila FR, Torres-Guzman RA, Maita K, Garcia JP, Serrano LP, Ho O, Forte AJ. Cell-Based Therapies Induce Tolerance of Vascularized Composite Allotransplants: A Systematic Review. J Surg Res 2024; 300:389-401. [PMID: 38851085 DOI: 10.1016/j.jss.2024.04.079] [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: 04/15/2023] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 06/10/2024]
Abstract
INTRODUCTION Vascularized composite allotransplantation (VCA) is the transplantation of multiple tissue types as a solution for devastating injuries. Despite the highly encouraging functional outcomes of VCA, the consequences of long-term immunosuppression remain the main obstacle in its application. In this review, we provide researchers and surgeons with a summary of the latest advances in the field of cell-based therapies for VCA tolerance. METHODS Four electronic databases were searched: PubMed, Scopus, Cumulative Index to Nursing and Allied Health Literature , and Web of Science. We used the Preferred Reporting Items for Systematic Reviews and Meta-Analysis as the basis of our organization. RESULTS Hematopoietic stem cells prolonged VCA survival. A combination of immature dendritic cells and tacrolimus was superior to tacrolimus alone. T cell Ig domain and mucin domain modified mature dendritic cells increased VCA tolerance. Bone marrow-derived mesenchymal stem cells prolonged survival of VCAs. A combination of adipose-derived mesenchymal stem cells, cytotoxic T-lymphocyte antigen 4 immunoglobulin, and antilymphocyte serum significantly improved VCA tolerance. Ex-vivo allotransplant perfusion with recipient's bone marrow-derived mesenchymal stem cells increased VCA survival. Recipient's adipose-derived mesenchymal stem cells and systemic immunosuppression prolonged VCA survival more than any of those agents alone. Additionally, a combination of peripheral blood mononuclear cells shortly incubated in mitomycin and cyclosporine significantly improved VCA survival. Finally, a combination of donor recipient chimeric cells, anti-αβ-T cell receptor (TCR), and cyclosporine significantly prolonged VCA tolerance. CONCLUSIONS Evidence from animal studies shows that cell-based therapies can prolong survival of VCAs. However, there remain many obstacles for these therapies, and they require rigorous clinical research given the rarity of the subjects and the complexity of the therapies. The major limitations of cell-based therapies include the need for conditioning with immunosuppressive drugs and radiation, causing significant toxicity. Safety concerns also persist as most research is on animal models. While completely replacing traditional immunosuppression with cell-based methods is unlikely soon, these therapies could reduce the need for high doses of immunosuppressants and improve VCA tolerance.
Collapse
Affiliation(s)
| | | | | | - Karla Maita
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - John P Garcia
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | | | - Olivia Ho
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
7
|
Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products-a review. Front Cell Dev Biol 2024; 12:1400347. [PMID: 39129786 PMCID: PMC11310176 DOI: 10.3389/fcell.2024.1400347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Mesenchymal stromal stem cells (MSCs) possess a remarkable potential for numerous clinical applications due to their unique properties including self-renewal, immunomodulation, paracrine actions and multilineage differentiation. However, the translation of MSC-based Advanced Therapy Medicinal Products (ATMPs) into the clinic has frequently met with inconsistent outcomes. One of the suspected reasons for this issue is the inherent and extensive variability that exists among such ATMPs, which makes the interpretation of their clinical efficacy difficult to assess, as well as to compare the results of various studies. This variability stems from numerous reasons including differences in tissue sources, donor attributes, variances in manufacturing protocols, as well as modes of administration. MSCs can be isolated from various tissues including bone marrow, umbilical cord, adipose tissue and others, each with its unique phenotypic and functional characteristics. While MSCs from different sources do share common features, they also exhibit distinct gene expression profiles and functional properites. Donor-specific factors such as age, sex, body mass index, and underlying health conditions can influence MSC phenotype, morphology, differentiation potential and function. Moreover, variations in preparation of MSC products introduces additional heterogeneity as a result of cell culture media composition, presence or absence of added growth factors, use of different serum supplements and culturing techniques. Once MSC products are formulated, storage protocols play a pivotal role in its efficacy. Factors that affect cell viability include cell concentration, delivery solution and importantly, post-thawing protocols where applicable. Ensuing, differences in administration protocols can critically affect the distribution and functionallity of administered cells. As MSC-based therapies continue to advance through numerous clinical trials, implication of strategies to reduce product heterogeneity is imperative. Central to addressing these challenges is the need for precise prediction of clinical responses, which require well-defined MSC populations and harmonized assessment of their specific functions. By addressing these issues by meaningful approaches, such as, e.g., MSC pooling, the field can overcome barriers to advance towards more consistent and effective MSC-based therapies.
Collapse
Affiliation(s)
- Ana Bajc Česnik
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
| | - Urban Švajger
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
8
|
Sousa AR, Cunha AF, Santos-Coquillat A, Estrada BH, Spiller KL, Barão M, Rodrigues AF, Simões S, Vilaça A, Ferreira L, Oliveira MB, Mano JF. Shape-Versatile Fixed Cellular Materials for Multiple Target Immunomodulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405367. [PMID: 38739450 PMCID: PMC11272431 DOI: 10.1002/adma.202405367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Therapeutic cells are usually administered as living agents, despite the risks of undesired cell migration and acquisition of unpredictable phenotypes. Additionally, most cell-based therapies rely on the administration of single cells, often associated with rapid in vivo clearance. 3D cellular materials may be useful to prolong the effect of cellular therapies and offer the possibility of creating structural volumetric constructs. Here, the manufacturing of shape-versatile fixed cell-based materials with immunomodulatory properties is reported. Living cell aggregates with different shapes (spheres and centimeter-long fibers) are fixed using a method compatible with maintenance of structural integrity, robustness, and flexibility of 3D constructs. The biological properties of living cells can be modulated before fixation, rendering an in vitro anti-inflammatory effect toward human macrophages, in line with a decreased activation of the nuclear factor kappa B (NF-κB) pathway that preponderantly correlated with the surface area of the materials. These findings are further corroborated in vivo in mouse skin wounds. Contact with fixed materials also reduces the proliferation of activated primary T lymphocytes, while promoting regulatory populations. The fixation of cellular constructs is proposed as a versatile phenotypic stabilization method that can be easily implemented to prepare immunomodulatory materials with therapeutic potential.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana F Cunha
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana Santos-Coquillat
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Beatriz Hernaez Estrada
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Marta Barão
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Artur Filipe Rodrigues
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Susana Simões
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Andreia Vilaça
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Lino Ferreira
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
- FMUC-Faculty of Medicine, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
9
|
Kheder RK, Darweesh O, Hussen BM, Abdullah SR, Basiri A, Taheri M. Mesenchymal stromal cells (MSCs) as a therapeutic agent of inflammatory disease and infectious COVID-19 virus: live or dead mesenchymal? Mol Biol Rep 2024; 51:295. [PMID: 38340168 DOI: 10.1007/s11033-023-09174-x] [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: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/12/2024]
Abstract
The COVID-19 infection is a worldwide disease that causes numerous immune-inflammatory disorders, tissue damage, and lung dysfunction. COVID-19 vaccines, including those from Pfizer, AstraZeneca, and Sinopharm, are available globally as effective interventions for combating the disease. The severity of COVID-19 can be most effectively reduced by mesenchymal stromal cells (MSCs) because they possess anti-inflammatory activity and can reverse lung dysfunction. MSCs can be harvested from various sources, such as adipose tissue, bone marrow, peripheral blood, inner organs, and neonatal tissues. The regulation of inflammatory cytokines is crucial in inhibiting inflammatory diseases and promoting the presence of anti-inflammatory cytokines for infectious diseases. MSCs have been employed as therapeutic agents for tissue damage, diabetes, autoimmune diseases, and COVID-19 patients. Our research aimed to determine whether live or dead MSCs are more suitable for the treatment of COVID-19 patients. Our findings concluded that dead MSCs, when directly administered to the patient, offer advantages over viable MSCs due to their extended presence and higher levels of immune regulation, such as T-reg, B-reg, and IL-10, compared to live MSCs. Additionally, dead and apoptotic MSCs are likely to be more readily captured by monocytes and macrophages, prolonging their presence compared to live MSCs.
Collapse
Affiliation(s)
- Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Sulaymaniyah, Iraq
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Omeed Darweesh
- College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Erbil, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Scineces, Tehran, Iran
| | - Mohammad Taheri
- Institue of Human Genetics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
10
|
Singh AK, Prasad P, Cancelas JA. Mesenchymal stromal cells, metabolism, and mitochondrial transfer in bone marrow normal and malignant hematopoiesis. Front Cell Dev Biol 2023; 11:1325291. [PMID: 38169927 PMCID: PMC10759248 DOI: 10.3389/fcell.2023.1325291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Hematopoietic stem cell (HSC) transplantation-based treatments are in different phases of clinical development, ranging from current therapies to a promise in the repair and regeneration of diseased tissues and organs. Mesenchymal stromal/stem cells (MSCs), which are fibroblast-like heterogeneous progenitors with multilineage differentiation (osteogenic, chondrogenic, and adipogenic) and self-renewal potential, and exist in the bone marrow (BM), adipose, and synovium, among other tissues, represent one of the most widely used sources of stem cells in regenerative medicine. MSCs derived from bone marrow (BM-MSCs) exhibit a variety of traits, including the potential to drive HSC fate and anti-inflammatory and immunosuppressive capabilities via paracrine activities and interactions with the innate and adaptive immune systems. The role of BM-MSC-derived adipocytes is more controversial and may act as positive or negative regulators of benign or malignant hematopoiesis based on their anatomical location and functional crosstalk with surrounding cells in the BM microenvironment. This review highlights the most recent clinical and pre-clinical findings on how BM-MSCs interact with the surrounding HSCs, progenitors, and immune cells, and address some recent insights on the mechanisms that mediate MSCs and adipocyte metabolic control through a metabolic crosstalk between BM microenvironment cells and intercellular mitochondrial transfer in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
11
|
Lin R, Zhang T, Gao J. Apoptotic Vesicles of MSCs: The Natural Therapeutic Agents and Bio-Vehicles for Targeting Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301671. [PMID: 37491784 DOI: 10.1002/smll.202301671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/11/2023] [Indexed: 07/27/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapies are increasingly recognized as promising cellular therapeutics and show the ability to treat various diseases. However, the underlying mechanism is not fully elucidated. Some recent studies have shown an unexpected result whereby MSCs undergo rapid apoptosis following administration but still exert therapeutic effects in some disease treatments. Such a therapeutic mechanism is believed to associate with the released apoptotic vesicles from apoptotic MSCs (MSC-ApoVs). This finding inspires a novel therapeutic strategy for using MSC-ApoVs for disease treatment. The present review aims to summarize the biogenesis, physiological functions, therapeutic potentials, and related mechanisms of apoptotic vesicles in MSC-based therapy. In addition, the potential applications of MSC-ApoVs as natural therapeutic agents and natural drug delivery vehicles are proposed and highlighted. The present review is hoped to provide a general understanding of MSC-ApoVs in disease treatment and inspire potential applications in targeted drug delivery.
Collapse
Affiliation(s)
- Ruyi Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Department of Pharmacy, Ningbo First Hospital, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| |
Collapse
|
12
|
da Silva TB, Rendra E, David CAW, Bieback K, Cross MJ, Wilm B, Liptrott NJ, Murray P. Umbilical cord mesenchymal stromal cell-derived extracellular vesicles lack the potency to immunomodulate human monocyte-derived macrophages in vitro. Biomed Pharmacother 2023; 167:115624. [PMID: 37783151 DOI: 10.1016/j.biopha.2023.115624] [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: 07/01/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been reported to display efficacy in a variety of preclinical models, but without long-term engraftment, suggesting a role for secreted factors, such as MSC-derived extracellular vesicles (EVs). MSCs are known to elicit immunomodulatory effects, an important aspect of which is their ability to affect macrophage phenotype. However, it is not clear if these effects are mediated by MSC-derived EVs, or other factors secreted by the MSCs. Here, we use flow cytometry to assess the effects of human umbilical cord (hUC) MSC-derived EVs on the expression of pro-inflammatory (CD80) and anti-inflammatory (CD163) surface markers in human monocyte-derived macrophages (hMDMs). hUC-MSC-derived EVs did not change the surface marker expression of the hMDMs. In contrast, when hMDMs were co-incubated with hUC-MSCs in indirect co-cultures, changes were observed in the expression of CD14, CD80 and CD163, particularly in M1 macrophages, suggesting that soluble factors are necessary to elicit a shift in phenotype. However, even though EVs did not alter the surface marker expression of macrophages, they promoted angiogenesis and phagocytic capacity increased proportionally to increases in EV concentration. Taken together, these results suggest that hUC-MSC-derived EVs are not sufficient to alter macrophage phenotype and that additional MSC-derived factors are needed.
Collapse
Affiliation(s)
- Tamiris Borges da Silva
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK
| | - Erika Rendra
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Christopher A W David
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Michael J Cross
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK.
| |
Collapse
|
13
|
Hernandez Pichardo A, Wilm B, Liptrott NJ, Murray P. Intravenous Administration of Human Umbilical Cord Mesenchymal Stromal Cells Leads to an Inflammatory Response in the Lung. Stem Cells Int 2023; 2023:7397819. [PMID: 37705699 PMCID: PMC10497368 DOI: 10.1155/2023/7397819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/25/2023] [Accepted: 08/04/2023] [Indexed: 09/15/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) administered intravenously (IV) have shown efficacy in preclinical models of various diseases. This is despite the cells not reaching the site of injury due to entrapment in the lungs. The immunomodulatory properties of MSCs are thought to underlie their therapeutic effects, irrespective of whether they are sourced from bone marrow, adipose tissue, or umbilical cord. To better understand how MSCs affect innate immune cell populations in the lung, we evaluated the distribution and phenotype of neutrophils, monocytes, and macrophages by flow cytometry and histological analyses after delivering human umbilical cord-derived MSCs (hUC-MSCs) IV into immunocompetent mice. After 2 hr, we observed a significant increase in neutrophils, and proinflammatory monocytes and macrophages. Moreover, these immune cells localized in close proximity to the MSCs, suggesting an active role in their clearance. By 24 hr, we detected an increase in anti-inflammatory monocytes and macrophages. These results suggest that the IV injection of hUC-MSCs leads to an initial inflammatory phase in the lung shortly after injection, followed by a resolution phase 24 hr later.
Collapse
Affiliation(s)
- Alejandra Hernandez Pichardo
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Neill J. Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
14
|
Lu D, Jiao X, Jiang W, Yang L, Gong Q, Wang X, Wei M, Gong S. Mesenchymal stem cells influence monocyte/macrophage phenotype: Regulatory mode and potential clinical applications. Biomed Pharmacother 2023; 165:115042. [PMID: 37379639 DOI: 10.1016/j.biopha.2023.115042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from a variety of tissues, such as umbilical cord, fat, and bone marrow. Today, MSCs are widely recognized for their prominent anti-inflammatory properties in a variety of acute and chronic inflammatory diseases. In inflammatory diseases, monocytes/macrophages are an important part of the innate immune response in the body, and the alteration of the inflammatory phenotype plays a crucial role in the secretion of pro-inflammatory/anti-inflammatory factors, the repair of injured sites, and the infiltration of inflammatory cells. In this review, starting from the effect of MSCs on the monocyte/macrophage phenotype, we have outlined in detail the process by which MSCs influence the transformation of the monocyte/macrophage inflammatory phenotype, emphasizing the central role of monocytes/macrophages in MSC-mediated anti-inflammatory and damage site repair. MSCs are phagocytosed by monocytes/macrophages in various physiological states, the paracrine effect of MSCs and mitochondrial transfer of MSCs to macrophages to promote the transformation of monocytes/macrophages into anti-inflammatory phenotypes. We also review the clinical applications of the MSCs-monocytes/macrophages system and describe novel pathways between MSCs and tissue repair, the effects of MSCs on the adaptive immune system, and the effects of energy metabolism levels on monocyte/macrophage phenotypic changes.
Collapse
Affiliation(s)
- Dejin Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xue Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Wenjian Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Li Yang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qian Gong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xiaobin Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shiqiang Gong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
15
|
Schrodt MV, Behan-Bush RM, Liszewski JN, Humpal-Pash ME, Boland LK, Scroggins SM, Santillan DA, Ankrum JA. Efferocytosis of viable versus heat-inactivated MSC induces human monocytes to distinct immunosuppressive phenotypes. Stem Cell Res Ther 2023; 14:206. [PMID: 37592321 PMCID: PMC10433682 DOI: 10.1186/s13287-023-03443-z] [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: 02/23/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Immunomodulation by mesenchymal stromal cells (MSCs) can occur through trophic factor mechanisms, however, intravenously infused MSCs are rapidly cleared from the body yet a potent immunotherapeutic response is still observed. Recent work suggests that monocytes contribute to the clearance of MSCs via efferocytosis, the body's natural mechanism for clearing dead and dying cells in a non-inflammatory manner. This begs the questions of how variations in MSC quality affect monocyte phenotype and if viable MSCs are even needed to elicit an immunosuppressive response. METHODS Herein, we sought to dissect MSC's trophic mechanism from their efferocytic mechanisms and determine if the viability of MSCs prior to efferocytosis influences the resultant phenotype of monocytes. We cultured viable or heat-inactivated human umbilical cord MSCs with human peripheral blood mononuclear cells for 24 h and observed changes in monocyte surface marker expression and secretion profile. To isolate the effect of efferocytosis from MSC trophic factors, we used cell separation techniques to remove non-efferocytosed MSCs before challenging monocytes to suppress T-cells or respond to inflammatory stimuli. For all experiments, viable and heat-inactivated efferocytic-licensing of monocytes were compared to non-efferocytic-licensing control. RESULTS We found that monocytes efferocytose viable and heat-inactivated MSCs equally, but only viable MSC-licensed monocytes suppress activated T-cells and suppression occurred even after depletion of residual MSCs. This provides direct evidence that monocytes that efferocytose viable MSCs are immunosuppressive. Further characterization of monocytes after efferocytosis showed that uptake of viable-but not heat inactivated-MSC resulted in monocytes secreting IL-10 and producing kynurenine. When monocytes were challenged with LPS, IL-2, and IFN-γ to simulate sepsis, monocytes that had efferocytosed viable MSC had higher levels of IDO while monocytes that efferocytosed heat inactivated-MSCs produced the lowest levels of TNF-α. CONCLUSION Collectively, these studies show that the quality of MSCs efferocytosed by monocytes polarize monocytes toward distinctive immunosuppressive phenotypes and highlights the need to tailor MSC therapies for specific indications.
Collapse
Affiliation(s)
- Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Riley M Behan-Bush
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Jesse N Liszewski
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Madeleine E Humpal-Pash
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Lauren K Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Sabrina M Scroggins
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA.
- , 103 S. Capitol St., 5621 SC, Iowa City, IA, 52242, USA.
| |
Collapse
|
16
|
Aabling RR, Alstrup T, Kjær EM, Poulsen KJ, Pedersen JO, Revenfeld AL, Møller BK, Eijken M. Reconstitution and post-thaw storage of cryopreserved human mesenchymal stromal cells: Pitfalls and optimizations for clinically compatible formulants. Regen Ther 2023; 23:67-75. [PMID: 37153832 PMCID: PMC10154666 DOI: 10.1016/j.reth.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/23/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction The regenerative and immunomodulatory properties of multipotent mesenchymal stromal cells (MSCs) make them an intriguing asset for therapeutic applications. An off-the-shelf approach, using pre-expanded cryopreserved allogenic MSCs, bypasses many practical difficulties of cellular therapy. Reconstitution of a MSC product away from cytotoxic cryoprotectants towards a preferred administration solution might be favorable for several indications. Variations in MSC handling accompanied by a non-standardized use of reconstitution solutions complicate a general clinical standardization of MSC cellular therapies. In this study, we aimed to identify a simple and clinically compatible approach for thawing, reconstitution, and post-thaw storage of cryopreserved MSCs. Methods Human adipose tissue-derived MSCs were expanded in human platelet lysate (hPL) supplemented culture medium and cryopreserved using a dimethyl sulfoxide (DMSO)-based cryoprotectant. Isotonic solutions (saline, Ringer's acetate and phosphate buffered saline (PBS)) with or without 2% human serum albumin (HSA) were used as thawing, reconstitution, and storage solutions. MSCs were reconstituted to 5 × 106 MSCs/mL for evaluating MSC stability. Total MSC numbers and viability were determined using 7-aminoactinomycin D (7-AAD) and flow cytometry. Results For thawing cryopreserved MSCs the presence of protein was proven to be essential. Up to 50% of MSCs were lost when protein-free thawing solutions were used. Reconstitution and post-thaw storage of MSCs in culture medium and widely used PBS demonstrated poor MSC stability (>40% cell loss) and viability (<80%) after 1 h of storage at room temperature. Reconstitution in simple isotonic saline appeared to be a good alternative for post-thaw storage, ensuring >90% viability with no observed cell loss for at least 4 h. Reconstitution of MSCs to low concentrations was identified as critical. Diluting MSCs to <105/mL in protein-free vehicles resulted in instant cell loss (>40% cell loss) and lower viability (<80%). Addition of clinical grade HSA could prevent cell loss during thawing and dilution. Conclusion This study identified a clinically compatible method for MSC thawing and reconstitution that ensures high MSC yield, viability, and stability. The strength of the method lies within the simplicity of implementation which offers an accessible way to streamline MSC therapies across different laboratories and clinical trials, improving standardization in this field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marco Eijken
- Corresponding author. Center of Gene and Cellular Therapy, Department of Clinical Immunology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark.
| |
Collapse
|
17
|
Shou Y, Liu L, Liu Q, Le Z, Lee KL, Li H, Li X, Koh DZ, Wang Y, Liu TM, Yang Z, Lim CT, Cheung C, Tay A. Mechano-responsive hydrogel for direct stem cell manufacturing to therapy. Bioact Mater 2023; 24:387-400. [PMID: 36632503 PMCID: PMC9817177 DOI: 10.1016/j.bioactmat.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin β1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this 'all-in-one' technology could offer a paradigm shift to existing standards in MSC therapy.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| | - Qimin Liu
- School of Civil Engineering and Architecture, Wuhan University of Technology, 430070, Wuhan, China
| | - Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Hua Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Dion Zhanyun Koh
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Yuwen Wang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
18
|
Wu HW, Chen HD, Chen YH, Mao XL, Feng YY, Li SW, Zhou XB. The Effects of Programmed Cell Death of Mesenchymal Stem Cells on the Development of Liver Fibrosis. Stem Cells Int 2023; 2023:4586398. [PMID: 37214784 PMCID: PMC10195177 DOI: 10.1155/2023/4586398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/06/2023] [Accepted: 04/02/2023] [Indexed: 05/24/2023] Open
Abstract
Mesenchymal stem cells have shown noticeable potential for unlimited self-renewal. They can differentiate into specific somatic cells, integrate into target tissues via cell-cell contact, paracrine effects, exosomes, and other processes and then regulate the target cells and tissues. Studies have demonstrated that transplantation of MSCs could decrease the expression and concentration of collagen in the liver, thereby reducing liver fibrosis. A growing body of evidence indicates that apoptotic MSCs could inhibit harmful immune responses and reduce inflammatory responses more effectively than viable MSCs. Accumulating evidence suggests that mitochondrial transfer from MSCs is a novel strategy for the regeneration of various damaged cells via the rescue of their respiratory activities. This study is aimed at reviewing the functions of MSCs and the related roles of the programmed cell death of MSCs, including autophagy, apoptosis, pyroptosis, and ferroptosis, as well as the regulatory pathogenic mechanisms of MSCs in liver fibrosis. Research has demonstrated that the miR-200B-3p gene is differentially expressed gene between LF and normal liver samples, and that the miR-200B-3p gene expression is positively correlated with the degree of liver fibrosis, suggesting that MSCs could inhibit liver fibrosis through pyroptosis. It was confirmed that circulating monocytes could deliver MSC-derived immunomodulatory molecules to different sites by phagocytosis of apoptotic MSCs, thereby achieving systemic immunosuppression. Accordingly, it was suggested that characterization of the programmed cell death-mediated immunomodulatory signaling pathways in MSCs should be a focus of research.
Collapse
Affiliation(s)
- Hong-wei Wu
- Department of Infectious Diseases, Taizhou Enze Medical Center (Group) Enze Hospital, Taizhou, Zhejiang, China
| | - He-dan Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ya-hong Chen
- Health Management Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xin-li Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yu-yi Feng
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shao-wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xian-bin Zhou
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
19
|
Wanyan P, Wang X, Li N, Huang Y, She Y, Zhang L. Mesenchymal stem cells therapy for acute kidney injury: A systematic review with meta-analysis based on rat model. Front Pharmacol 2023; 14:1099056. [PMID: 37124211 PMCID: PMC10133560 DOI: 10.3389/fphar.2023.1099056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Objective: To systematically evaluate the efficacy of mesenchymal stem cells (MSCs) for acute kidney injury (AKI) in preclinical studies and to explore the optimal transplantation strategy of MSCs by network meta-analysis with the aim of improving the efficacy of stem cell therapy. Methods: Computer searches of PubMed, Web of Science, Cochrane, Embase, CNKI, Wanfang, VIP, and CBM databases were conducted until 17 August 2022. Literature screening, data extraction and quality evaluation were performed independently by two researchers. Results and Discussion: A total of 50 randomized controlled animal studies were included. The results of traditional meta-analysis showed that MSCs could significantly improve the renal function and injured renal tissue of AKI rats in different subgroups. The results of network meta-analysis showed that although there was no significant difference in the therapeutic effect between different transplant routes and doses of MSCs, the results of surface under the cumulative ranking probability curve (SUCRA) showed that the therapeutic effect of intravenous transplantation of MSCs was better than that of arterial and intrarenal transplantation, and the therapeutic effect of high dose (>1×106) was better than that of low dose (≤1×106). However, the current preclinical studies have limitations in experimental design, measurement and reporting of results, and more high-quality studies, especially direct comparative evidence, are needed in the future to further confirm the best transplantation strategy of MSCs in AKI. Systematic Review Registration: identifier https://CRD42022361199, https://www.crd.york.ac.uk/prospero.
Collapse
Affiliation(s)
- Pingping Wanyan
- Department of Pathology and Pathophysiology, School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xin Wang
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, China
- Department of Surgery, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Nenglian Li
- Department of Pathology and Pathophysiology, School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yong Huang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yali She
- Department of Pathology and Pathophysiology, School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Li Zhang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
20
|
Ercelen N, Karasu N, Kahyaoglu B, Cerezci O, Akduman RC, Ercelen D, Erturk G, Gulay G, Alpaydin N, Boyraz G, Monteleone B, Kural Z, Silek H, Temur S, Bingol CA. Clinical experience: Outcomes of mesenchymal stem cell transplantation in five stroke patients. Front Med (Lausanne) 2023; 10:1051831. [PMID: 36744151 PMCID: PMC9892908 DOI: 10.3389/fmed.2023.1051831] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Stem cell therapy, which has promising results in acute disorders such as stroke, supports treatment by providing rehabilitation in the chronic stage patients. In acute stroke, thrombolytic medical treatment protocols are clearly defined in neurologic emergencies, but in neurologic patients who miss the "thrombolytic treatment intervention window," or in cases of hypoxic-ischemic encephalopathy, our hands are tied, and we are still unfortunately faced with hopeless clinical implementations. We consider mesenchymal stem cell therapy a viable option in these cases. In recent years, novel research has focused on neuro-stimulants and supportive and combined therapies for stroke. Currently, available treatment options are limited, and only certain patients are eligible for acute treatment. In the scope of our experience, five stroke patients were evaluated in this study, who was treated with a single dose of 1-2 × 106 cells/kg allogenic umbilical cord-mesenchymal stem cells (UC-MSCs) with the official confirmation of the Turkish Ministry of Health Stem Cell Commission. The patients were followed up for 12 months, and clinical outcomes are recorded. NIH Stroke Scale/Scores (NIHSS) decreased significantly (p = 0.0310), and the Rivermead Assessment Scale (RMA) increased significantly (p = 0.0234) for all patients at the end of the follow-up. All the patients were followed up for 1 year within a rehabilitation program. Major clinical outcome improvements were observed in the overall clinical conditions of the UC-MSC treatment patients. We observed improvement in the patients' upper extremity and muscle strength, spasticity, and fine motor functions. Considering recent studies in the literature together with our results, allogenic stem cell therapies are introduced as promising novel therapies in terms of their encouraging effects on physiological motor outcomes.
Collapse
Affiliation(s)
- Nesrin Ercelen
- Department of Medical Genetics, Faculty of Medicine, Üsküdar University, Istanbul, Türkiye,*Correspondence: Nesrin Ercelen,
| | - Nilgun Karasu
- Department of Medical Genetics, Faculty of Medicine, Üsküdar University, Istanbul, Türkiye
| | | | - Onder Cerezci
- Department of Physical Therapy and Rehabilitation, Faculty of Medicine, Üsküdar University, Istanbul, Türkiye,Department of Physical Medicine and Rehabilitation, American Hospital, Istanbul, Türkiye
| | - Rana Cagla Akduman
- Department of Neurology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Defne Ercelen
- Computational and Systems Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gizem Erturk
- Department of Neurology, American Hospital, Istanbul, Türkiye,Department of Healthcare Management, Faculty of Health Sciences, Üsküdar University, Istanbul, Türkiye
| | - Gokay Gulay
- ATIGEN-CELL/Cell and Tissue Center, Trabzon, Türkiye
| | | | - Gizem Boyraz
- Geneis Genetic System Solutions, Istanbul, Türkiye
| | - Berrin Monteleone
- Department of Pediatrics at NYU Long Island School of Medicine, Medical Genetics, Langone Hospital, New York University, Long Island, NY, United States
| | - Zekiye Kural
- Department of Neurology, American Hospital, Istanbul, Türkiye
| | - Hakan Silek
- Department of Neurology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Sibel Temur
- Department of Anesthesia and Reanimation, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Canan Aykut Bingol
- Department of Neurology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| |
Collapse
|
21
|
Cheng J. Cell-Based Therapies in Clinical Pain Management. NEUROIMMUNE INTERACTIONS IN PAIN 2023:273-286. [DOI: 10.1007/978-3-031-29231-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
22
|
Kholodenko IV, Kholodenko RV, Majouga AG, Yarygin KN. Apoptotic MSCs and MSC-Derived Apoptotic Bodies as New Therapeutic Tools. Curr Issues Mol Biol 2022; 44:5153-5172. [PMID: 36354663 PMCID: PMC9688732 DOI: 10.3390/cimb44110351] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 12/03/2022] Open
Abstract
Over the past two decades, mesenchymal stem cells (MSCs) have shown promising therapeutic effects both in preclinical studies (in animal models of a wide range of diseases) and in clinical trials. However, the efficacy of MSC-based therapy is not always predictable. Moreover, despite the large number of studies, the mechanisms underlying the regenerative potential of MSCs are not fully elucidated. Recently, it has been reliably established that transplanted MSCs can undergo rapid apoptosis and clearance from the recipient's body, still exhibiting therapeutic effects, especially those associated with their immunosuppressive/immunomodulating properties. The mechanisms underlying these effects can be mediated by the efferocytosis of apoptotic MSCs by host phagocytic cells. In this concise review, we briefly describe three types of MSC-generated extracellular vesicles, through which their therapeutic functions can potentially be carried out; we focused on reviewing recent data on apoptotic MSCs and MSC-derived apoptotic bodies (MSC-ApoBDs), their functions, and the mechanisms of their therapeutic effects.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| |
Collapse
|
23
|
Owen A, Patten D, Vigneswara V, Frampton J, Newsome PN. PDGFRα/Sca-1 Sorted Mesenchymal Stromal Cells Reduce Liver Injury in Murine Models of Hepatic Ischemia-Reperfusion Injury. Stem Cells 2022; 40:1056-1070. [PMID: 35999023 PMCID: PMC9707286 DOI: 10.1093/stmcls/sxac059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/06/2022] [Indexed: 11/12/2022]
Abstract
Liver transplantation is an effective therapy, but increasing demand for donor organs has led to the use of marginal donor organs with increased complication rates. Mesenchymal stromal cells (MSC) pleiotropically modulate aberrant immune-mediated responses and represent a potential therapy to target the inflammation seen post-transplant with marginal donor livers. To avoid the confounding effects of xenotransplantation seen in studies with human MSC, a PDGFRα/Sca-1 (PaS) sorted MSC population was used which was analogous to human MSC populations (LNGFR+Thy-1+VCAM-1Hi). PaS MSC are a well-described population that demonstrate MSC properties without evidence of clonal mutation during expansion. We demonstrate their anti-inflammatory properties herein through their suppression of T-lymphocyte proliferation in vitro and secretion of anti-inflammatory cytokines (IL-10 and OPG) after stimulation (P = .004 and P = .003). The MDR2-/- model of biliary injury and hepatic ischemia-reperfusion (HIR) injury models were used to replicate the non-anastomotic biliary complications seen following liver transplantation. Systemic MSC therapy in MDR2-/- mice led to reduced liver injury with an increase in restorative macrophages (5913 ± 333.9 vs 12 597 ± 665.8, P = .002, n = 7) and a change in lymphocyte ratios (3.55 ± 0.37 vs 2.59 ± 0.139, P = .023, n = 17), whereas subcutaneous administration of MSC showed no beneficial effect. MSC also reduced cell death in the HIR model assessed by Periodic acid-Schiff (PAS) staining (91.7% ± 2.8 vs 80.1% ± 4.6, P = .03). Systemically administered quantum dot-labeled MSC were tracked using single-cell resolution CryoViz imaging which demonstrated their sequestration in the lungs alongside retention/redistribution to injured liver tissue. MSC represent a potential novel therapy in marginal organ transplantation which warrants further study.
Collapse
Affiliation(s)
| | | | | | | | - Philip N Newsome
- Corresponding author: Philip N. Newsome, Centre for Liver and Gastrointestinal Research, Institute of Biomedical Research, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK.
| |
Collapse
|
24
|
Surtaieva YV, Mazurkevich AY, Bokotko RR. Effects of transplanted mesenchymal stem cells on repair of the lung tissue of rats with experimental pulmonary fibrosis. REGULATORY MECHANISMS IN BIOSYSTEMS 2022. [DOI: 10.15421/022240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pulmonary fibrosis is one of the commonest forms of interstitial lung diseases with poorly studied methods of its treatment in both human and veterinary medicines. Therefore, this paper focused on seeking alternative methods of its diagnostics and treatment. The article provides the results of the study of bronchoalveolar lavage fluid of rats with experimental lung fibrosis and influence of transplanted allogeneic mesenchymal stem cells of the bone marrow on stimulation of regenerative processes in damaged lung tissues. The studies were conducted on female Wistar rats with pulmonary fibrosis modeled using single transthoracic injection of solution of bleomycin hydrochloride. For the purpose of treatment, we used allogeneic mesenchymal stem cells introduced by various methods and the traditional treatment. We determined that best normalization of the parameters of the studied brochoalveolar lavage occurred in animals that received mesenchymal stem cells. The most active repair processes were in the experimental group that received the mesenchymal stem cells directly to the lung tissue. The animals that received intravenous injection of mesenchymal stemm cells were observed to have lower clinical parameters of the brochoalveolar lavage, but still better than such in the group treated traditionally. The lowest parameters were in animals that received the traditional treatment; they were greater than the phisological parameters, but significantly exceeded them in animals of the control group, indicating presence of inflammatory process in the lung tissue. The conducted cytological assays of the samples of the brochoalveolar lavage revealed that experimental animals with experimental pulmonary fibrosis had development of macrophage and lymphocytic reactions under the influence of transplanted mesenchymal stemm cells. We observed no atypical cells in all the experimental groups. This allows us to draw a conclusion that using stem cells by various methods of transplantation does not stimulate the onset of negative reactons (formation of atypical cells, metastatic processes, etc). Thus, the results of the study of the influence of transplanted mesenchymal stem cells demonstrate that in the conditions of experimental pulmonary fibrosis, the activity of regenerative processes in pathologically altered lung tissue may be an effective method of treatment of animals with this kind of pathology.
Collapse
|
25
|
Zheng D, Bhuvan T, Payne NL, Heng TSP. Secondary Lymphoid Organs in Mesenchymal Stromal Cell Therapy: More Than Just a Filter. Front Immunol 2022; 13:892443. [PMID: 35784291 PMCID: PMC9243307 DOI: 10.3389/fimmu.2022.892443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic potential in inflammatory models of human disease. However, clinical translation has fallen short of expectations, with many trials failing to meet primary endpoints. Failure to fully understand their mechanisms of action is a key factor contributing to the lack of successful commercialisation. Indeed, it remains unclear how the long-ranging immunomodulatory effects of MSCs can be attributed to their secretome, when MSCs undergo apoptosis in the lung shortly after intravenous infusion. Their apoptotic fate suggests that efficacy is not based solely on their viable properties, but also on the immune response to dying MSCs. The secondary lymphoid organs (SLOs) orchestrate immune responses and play a key role in immune regulation. In this review, we will discuss how apoptotic cells can modify immune responses and highlight the importance of MSC-immune cell interactions in SLOs for therapeutic outcomes.
Collapse
Affiliation(s)
- Di Zheng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tejasvini Bhuvan
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Natalie L. Payne
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Tracy S. P. Heng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, Australia
- *Correspondence: Tracy S. P. Heng,
| |
Collapse
|
26
|
Srinivasan A, Sathiyanathan P, Yin L, Liu TM, Lam A, Ravikumar M, Smith RAA, Loh HP, Zhang Y, Ling L, Ng SK, Yang YS, Lezhava A, Hui J, Oh S, Cool SM. Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022; 24:456-472. [PMID: 35227601 DOI: 10.1016/j.jcyt.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Therapies using mesenchymal stromal cells (MSCs) to treat immune and inflammatory conditions are now at an exciting stage of development, with many MSC-based products progressing to phase II and III clinical trials. However, a major bottleneck in the clinical translation of allogeneic MSC therapies is the variable immunomodulatory properties of MSC products due to differences in their tissue source, donor heterogeneity and processes involved in manufacturing and banking. This variable functionality of MSC products likely contributes to the substantial inconsistency observed in the clinical outcomes of phase III trials of MSC therapies; several trials have failed to reach the primary efficacy endpoint. In this review, we discuss various strategies to consistently maintain or enhance the immunomodulatory potency of MSCs during ex vivo expansion, which will enable the manufacture of allogeneic MSC banks that have high potency and low variability. Biophysical and biochemical priming strategies, the use of culture additives such as heparan sulfates, and genetic modification can substantially enhance the immunomodulatory properties of MSCs during in vitro expansion. Furthermore, robust donor screening, the use of biomarkers to select for potent MSC subpopulations, and rigorous quality testing to improve the release criteria for MSC banks have the potential to reduce batch-to-batch heterogeneity and enhance the clinical efficacy of the final MSC product. Machine learning approaches to develop predictive models of individual patient response can enable personalized therapies and potentially establish correlations between in vitro potency measurements and clinical outcomes in human trials.
Collapse
Affiliation(s)
- Akshaya Srinivasan
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Lu Yin
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Alan Lam
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Maanasa Ravikumar
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Han Ping Loh
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Ying Zhang
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Ling Ling
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute, A*STAR, Singapore
| | | | - Alexander Lezhava
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - James Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR, Singapore.
| | - Simon M Cool
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
27
|
Mönch D, Reinders MEJ, Dahlke MH, Hoogduijn MJ. How to Make Sense out of 75,000 Mesenchymal Stromal Cell Publications? Cells 2022; 11:cells11091419. [PMID: 35563725 PMCID: PMC9101744 DOI: 10.3390/cells11091419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stromal cells have been the subject of an expanding number of studies over the past decades. Today, over 75,000 publications are available that shine light on the biological properties and therapeutic effects of these versatile cells in numerous pre-clinical models and early-phase clinical trials. The massive number of papers makes it hard for researchers to comprehend the whole field, and furthermore, they give the impression that mesenchymal stromal cells are wonder cells that are curative for any condition. It is becoming increasingly difficult to dissect how and for what conditions mesenchymal stromal cells exhibit true and reproducible therapeutic effects. This article tries to address the question how to make sense of 75,000, and still counting, publications on mesenchymal stromal cells.
Collapse
Affiliation(s)
- Dina Mönch
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany;
- University of Tübingen, 72074 Tübingen, Germany
| | - Marlies E. J. Reinders
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Marc H. Dahlke
- Department of Surgery, Robert-Bosch-Hospital, 70376 Stuttgart, Germany;
| | - Martin J. Hoogduijn
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
28
|
Fazekas B, Alagesan S, Watson L, Ng O, Conroy CM, Català C, Andres MV, Negi N, Gerlach JQ, Hynes SO, Lozano F, Elliman SJ, Griffin MD. Comparison of Single and Repeated Dosing of Anti-Inflammatory Human Umbilical Cord Mesenchymal Stromal Cells in a Mouse Model of Polymicrobial Sepsis. Stem Cell Rev Rep 2022; 18:1444-1460. [PMID: 35013938 PMCID: PMC8747454 DOI: 10.1007/s12015-021-10323-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs) ameliorate pre-clinical sepsis and sepsis-associated acute kidney injury (SA-AKI) but clinical trials of single-dose MSCs have not indicated robust efficacy. This study investigated immunomodulatory effects of a novel MSC product (CD362-selected human umbilical cord-derived MSCs [hUC-MSCs]) in mouse endotoxemia and polymicrobial sepsis models. Initially, mice received intra-peritoneal (i.p.) lipopolysaccharide (LPS) followed by single i.p. doses of hUC-MSCs or vehicle. Next, mice underwent cecal ligation and puncture (CLP) followed by intravenous (i.v.) doses of hUC-MSCs at 4 h or 4 and 28 h. Analyses included serum/plasma assays of biochemical indices, inflammatory mediators and the AKI biomarker NGAL; multi-color flow cytometry of peritoneal macrophages (LPS) and intra-renal immune cell subpopulations (CLP) and histology/immunohistochemistry of kidney (CLP). At 72 h post-LPS injections, hUC-MSCs reduced serum inflammatory mediators and peritoneal macrophage M1/M2 ratio. Repeated, but not single, hUC-MSC doses administered at 48 h post-CLP resulted in lower serum concentrations of inflammatory mediators, lower plasma NGAL and reversal of sepsis-associated depletion of intra-renal T cell and myeloid cell subpopulations. Hierarchical clustering analysis of all 48-h serum/plasma analytes demonstrated partial co-clustering of repeated-dose hUC-MSC CLP animals with a Sham group but did not reveal a distinct signature of response to therapy. It was concluded that repeated doses of CD362-selected hUC-MSCs are required to modulate systemic and local immune/inflammatory events in polymicrobial sepsis and SA-AKI. Inter-individual variability and lack of effect of single dose MSC administration in the CLP model are consistent with observations to date from early-phase clinical trials.
Collapse
Affiliation(s)
- Barbara Fazekas
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | | | - Olivia Ng
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Callum M Conroy
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Cristina Català
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Neema Negi
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Jared Q Gerlach
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
| | - Sean O Hynes
- Discipline of Pathology, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Histopathology, Galway University Hospitals, Galway, Ireland
| | - Francisco Lozano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Department de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | | | - Matthew D Griffin
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland.
- Department of Nephrology, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland.
- National University of Ireland Galway, REMEDI, Biomedical Sciences, Corrib Village, Dangan, Galway, H91 TK33, Ireland.
| |
Collapse
|
29
|
The Inability of Ex Vivo Expanded Mesenchymal Stem/Stromal Cells to Survive in Newborn Mice and to Induce Transplantation Tolerance. Stem Cell Rev Rep 2022; 18:2365-2375. [PMID: 35288846 DOI: 10.1007/s12015-022-10363-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 11/09/2022]
Abstract
An encounter of the developing immune system with an antigen results in the induction of immunological areactivity to this antigen. In the case of transplantation antigens, the application of allogeneic hematopoietic cells induces a state of neonatal transplantation tolerance. This tolerance depends on the establishment of cellular chimerism, when allogeneic cells survive in the neonatally treated recipient. Since mesenchymal stem/stromal cells (MSCs) have been shown to have low immunogenicity and often survive in allogeneic recipients, we attempted to use these cells for induction of transplantation tolerance. Newborn (less than 24 h old) C57BL/6 mice were injected intraperitoneally with 5 × 106 adipose tissue-derived MSCs isolated from allogeneic donors and the fate and survival of these cells were monitored. The impact of MSC application on the proportion of cell populations of the immune system and immunological reactivity was assessed. In addition, the survival of skin allografts in neonatally treated recipients was tested. We found that in vitro expanded MSCs did not survive in neonatal recipients, and the living MSCs were not detected few days after their application. Furthermore, there were no significant changes in the proportion of individual immune cell populations including CD4+ cell lineages, but we detected an apparent shift to the production of Th1 cytokines IL-2 and IFN-γ in neonatally treated mice. However, skin allografts in the MSC-treated recipients were promptly rejected. These results therefore show that in vitro expanded MSCs do not survive in neonatal recipients, but induce a cytokine imbalance without induction of transplantation tolerance.
Collapse
|
30
|
Mönch D, Koch J, Dahlke MH. Are Mesenchymal Stem Cells Fibroblasts with Benefits? CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00210-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Zulpaite R, Miknevicius P, Leber B, Strupas K, Stiegler P, Schemmer P. Ex-vivo Kidney Machine Perfusion: Therapeutic Potential. Front Med (Lausanne) 2021; 8:808719. [PMID: 35004787 PMCID: PMC8741203 DOI: 10.3389/fmed.2021.808719] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/06/2021] [Indexed: 01/11/2023] Open
Abstract
Kidney transplantation remains the gold standard treatment for patients suffering from end-stage kidney disease. To meet the constantly growing organ demands grafts donated after circulatory death (DCD) or retrieved from extended criteria donors (ECD) are increasingly utilized. Not surprisingly, usage of those organs is challenging due to their susceptibility to ischemia-reperfusion injury, high immunogenicity, and demanding immune regulation after implantation. Lately, a lot of effort has been put into improvement of kidney preservation strategies. After demonstrating a definite advantage over static cold storage in reduction of delayed graft function rates in randomized-controlled clinical trials, hypothermic machine perfusion has already found its place in clinical practice of kidney transplantation. Nevertheless, an active investigation of perfusion variables, such as temperature (normothermic or subnormothermic), oxygen supply and perfusate composition, is already bringing evidence that ex-vivo machine perfusion has a potential not only to maintain kidney viability, but also serve as a platform for organ conditioning, targeted treatment and even improve its quality. Many different therapies, including pharmacological agents, gene therapy, mesenchymal stromal cells, or nanoparticles (NPs), have been successfully delivered directly to the kidney during ex-vivo machine perfusion in experimental models, making a big step toward achievement of two main goals in transplant surgery: minimization of graft ischemia-reperfusion injury and reduction of immunogenicity (or even reaching tolerance). In this comprehensive review current state of evidence regarding ex-vivo kidney machine perfusion and its capacity in kidney graft treatment is presented. Moreover, challenges in application of these novel techniques in clinical practice are discussed.
Collapse
Affiliation(s)
- Ruta Zulpaite
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Povilas Miknevicius
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Bettina Leber
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | | | - Philipp Stiegler
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Peter Schemmer
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
32
|
SDF-1 α Facilitates Mesenchymal Stem Cells to Induce Regulatory B Cell Differentiation from Patients with Immune Thrombocytopenia. Stem Cells Int 2021; 2021:3254488. [PMID: 34790240 PMCID: PMC8592740 DOI: 10.1155/2021/3254488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2021] [Indexed: 11/28/2022] Open
Abstract
B cells play a central role in the pathogenesis of immune thrombocytopenia (ITP) by participating in humoral immunity. Meanwhile, regulatory B cells (Bregs), one subset of B cells, express negative regulatory effect on ITP. Mesenchymal stem cells (MSCs) have been demonstrated in the ability to induce immunosuppression, and stromal cell-derived factor-1α (SDF-1α) plays an important role in the migration and survival of MSCs. To investigate the mechanism of SDF-1α in controlling umbilical cord-derived MSCs (UC-MSCs) in inducing regulatory B cell differentiation of patients with ITP, we reconfirmed that SDF-1α promotes the proliferation of MSCs at the low doses of 0.05 μg/mL and 0.1 μg/mL but inhibits the proliferation and promotes the apoptosis of UC-MSCs at the high doses 0.5 μg/mL and 1 μg/mL; when UC-MSCs are cocultured with SDF-1α at 0.1 μg/mL, the decreased proportion of CD19+/CD24hi/CD38hi cells and IL-10-producing B cells (B 10 cell), considered as the Breg subset from ITP significantly enhanced, and the content of IL-10 in the supernatant is also obviously increased. The proportion of Bregs and the IL-10 secretion could be further promoted by the UC-MSCs treated with 0.1 μg/mL SDF-1α, which could also promote the miRNA-133 expression of UC-MSCs in an exosome-dependent manner; moreover, while the UC-MSCs were transfected with the miR-133 inhibitor, the proportion of induced Bregs decreased obviously when cocultured with peripheral blood mononuclear cells (PBMCs) of ITP. We conclude that UC-MSCs could effectively enhance the decreased proportion of Bregs from ITP; at appropriate concentrations, SDF-1α may promote the proliferating and survival ability of UC-MSCs and improve the production of Bregs induced by UC-MSCs through controlling miRNA-133 expression in the exosomes.
Collapse
|
33
|
Krampera M, Le Blanc K. Mesenchymal stromal cells: Putative microenvironmental modulators become cell therapy. Cell Stem Cell 2021; 28:1708-1725. [PMID: 34624232 DOI: 10.1016/j.stem.2021.09.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An exceptional safety profile has been shown in a large number of cell therapy clinical trials that use mesenchymal stromal cells (MSCs). However, reliable potency assays are still lacking to predict MSC immunosuppressive efficacy in the clinical setting. Nevertheless, MSCs are approved in Japan and Europe for the treatment of graft-versus-host and Crohn's fistular diseases, but not in the United States for any clinical indication. We discuss potential mechanisms of action for the therapeutic effects of MSC transplantation, experimental models that dissect tissue modulating function of MSCs, and approaches for identifying MSC effects in vivo by integrating biomarkers of disease and MSC activity.
Collapse
Affiliation(s)
- Mauro Krampera
- Section of Hematology and Bone Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy.
| | - Katarina Le Blanc
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Stockholm, Sweden; Center of Allogeneic Stem Cell Transplantation and Cellular Therapy (CAST), Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| |
Collapse
|
34
|
Sant'Ana AN, Araújo AB, Gonçalves FDC, Paz AH. Effects of living and metabolically inactive mesenchymal stromal cells and their derivatives on monocytes and macrophages. World J Stem Cells 2021; 13:1160-1176. [PMID: 34630856 PMCID: PMC8474715 DOI: 10.4252/wjsc.v13.i9.1160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/01/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewing stem cells that have great potential as cell therapy for autoimmune and inflammatory disorders, as well as for other clinical conditions, due to their immunoregulatory and regenerative properties. MSCs modulate the inflammatory milieu by releasing soluble factors and acting through cell-to-cell mechanisms. MSCs switch the classical inflammatory status of monocytes and macrophages towards a non-classical and anti-inflammatory phenotype. This is characterized by an increased secretion of anti-inflammatory cytokines, a decreased release of pro-inflammatory cytokines, and changes in the expression of cell membrane molecules and in metabolic pathways. The MSC modulation of monocyte and macrophage phenotypes seems to be critical for therapy effectiveness in several disease models, since when these cells are depleted, no immunoregulatory effects are observed. Here, we review the effects of living MSCs (metabolically active cells) and metabolically inactive MSCs (dead cells that lost metabolic activity by induced inactivation) and their derivatives (extracellular vesicles, soluble factors, extracts, and microparticles) on the profile of macrophages and monocytes and the implications for immunoregulatory and reparative processes. This review includes mechanisms of action exhibited in these different therapeutic approaches, which induce the anti-inflammatory properties of monocytes and macrophages. Finally, we overview several possibilities of therapeutic applications of these cells and their derivatives, with results regarding monocytes and macrophages in animal model studies and some clinical trials.
Collapse
Affiliation(s)
- Alexia Nedel Sant'Ana
- Laboratório de Células Tecidos e Genes, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Anelise Bergmann Araújo
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil.
| | | | - Ana Helena Paz
- Laboratório de Células Tecidos e Genes, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| |
Collapse
|
35
|
Nakai H, Fujita Y, Masuda S, Komatsu M, Tani A, Okita Y, Okada K, Kawamoto A. Intravenous injection of adult human bone marrow mesenchymal stromal cells attenuates spinal cord ischemia/reperfusion injury in a murine aortic arch crossclamping model. JTCVS OPEN 2021; 7:23-40. [PMID: 36003746 PMCID: PMC9390396 DOI: 10.1016/j.xjon.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 06/04/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE We sought to investigate the efficacy of human bone marrow mesenchymal stem/stromal cell (hBM-MSC) in a murine spinal cord ischemia/reperfusion (SCIR) model. METHODS C57BL/6J mice were subjected to SCIR by crossclamping the aortic arch and left subclavian artery for 5.5 minutes. Two hours after reperfusion, hBM-MSCs (hBM-MSC group) or phosphate-buffered saline (control group) were intravenously injected without immunosuppressant. Hindlimb motor function was assessed until day 28 after reperfusion using the Basso Mouse Scale (BMS). The lumbar spinal cord was harvested at hour 24 and day 28, and the histologic number of NeuN-positive motor neurons in 3 cross-sections of each lumbar spinal cord and the gene expression were evaluated. RESULTS BMS score was 0 throughout the study period in all control mice. BMS score was significantly greater in the hBM-MSC group than the control group from hour 8 (P < .05) to day 28 (P < .01). The numbers of motor neurons at hour 24 (P < .01) and day 28 (P < .05) were significantly preserved in the hBM-MSC group than the control group. mRNA expression levels of proinflammatory cytokines were significantly lower (P < .05), and those of insulin-like growth factor-1 (P < .01) and proangiogenic factors (P < .05) were significantly greater in the hBM-MSC group than the control group at hour 24. CONCLUSIONS hBM-MSC therapy may attenuate SCIR injury by preserving motor neurons, at least in part, through inhibition of proinflammatory cytokines and upregulation of proangiogenic factors in the reperfusion-injured spinal cord.
Collapse
Key Words
- BM, bone marrow
- BMS, Basso Mouse Scale
- EV, extracellular vesicle
- IGF-1, insulin-like growth factor-1
- IL-10, interleukin-10
- LSA, left subclavian artery
- PBS, phosphate-buffered saline
- SCI, spinal cord ischemia
- SCIR, spinal cord ischemia/reperfusion
- hBM-MSC, human bone marrow mesenchymal stem/stromal cell
- human bone marrow mesenchymal stromal cells
- mRNA, messenger RNA
- paraplegia
- spinal cord ischemia
- spinal cord reperfusion injury
- thoracic aortic surgery
Collapse
Affiliation(s)
- Hidekazu Nakai
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyuki Fujita
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Satoru Masuda
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Miki Komatsu
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Ayumi Tani
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Yutaka Okita
- Cardiovascular Center, Takatsuki General Hospital, Takatsuki, Japan
| | - Kenji Okada
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Atsuhiko Kawamoto
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
36
|
Storti G, Scioli MG, Kim BS, Terriaca S, Fiorelli E, Orlandi A, Cervelli V. Mesenchymal Stem Cells in Adipose Tissue and Extracellular Vesicles in Ovarian Cancer Patients: A Bridge toward Metastatic Diffusion or a New Therapeutic Opportunity? Cells 2021; 10:cells10082117. [PMID: 34440886 PMCID: PMC8392703 DOI: 10.3390/cells10082117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the deadliest malignancies among women. Approximately 75% of the patients with ovarian cancer are diagnosed with advanced disease that already has metastasis, particularly to the omentum. The omentum constitutes the ideal soil for ovarian cancer metastasis due to a complex intraperitoneal milieu that favors and supports the whole metastatic process. Adipose-derived stem/stromal cells (ADSCs) are part of this microenvironment and foster tumor progression via sustained paracrine secretion, including extracellular vesicles (EVs). Nonetheless, the preferential relationship between ADSCs, ADSC-derived EVs, and ovarian cancer cells could be exploited to use ADSCs and EVs as a vehicle for anti-cancer therapies. This review will analyze the strict relations between tumor progression, metastatic disease, and adipose tissue with its staminal components. In addition, we will describe the crosstalk and biologic relationship between ADSCs and tumor cells, the role of EVs in intercellular communication, the establishment of drug resistance, metastatic capacity, and ovarian cancer progression. We will analyze the new therapeutic opportunities in treating ovarian cancer offered by ADSCs and EVs as a vehicle for therapeutic molecules to target precisely tumor cells and limit the systemic adverse effects. Finally, we will discuss the limitations of these therapeutic approaches.
Collapse
Affiliation(s)
- Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy;
- Correspondence: ; Tel.: +39-06-23188514; Fax: +39-06-23188466
| | - Maria Giovanna Scioli
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Sonia Terriaca
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Elena Fiorelli
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy;
| |
Collapse
|
37
|
Carty F, Dunbar H, Hawthorne IJ, Ting AE, Stubblefield SR, Van't Hof W, English K. IFN-γ and PPARδ influence the efficacy and retention of multipotent adult progenitor cells in graft vs host disease. Stem Cells Transl Med 2021; 10:1561-1574. [PMID: 34397170 PMCID: PMC8550699 DOI: 10.1002/sctm.21-0008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/29/2021] [Accepted: 07/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cell‐based therapy for the treatment of inflammatory disorders has focused on the application of mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs). Despite the recent positive findings in industry‐sponsored clinical trials of MSCs and MAPCs for graft vs host disease (GvHD), cell therapy is efficacious in some but not all patients, highlighting the need to identify strategies to enhance cell‐based therapeutic efficacy. Here, we demonstrate the capacity for interferon (IFN)‐γ licensing to enhance human MAPC efficacy and retention following early administration in a humanized mouse model of acute GvHD (aGvHD). Activation of the nuclear receptor peroxisome proliferator‐activated receptor delta (PPARδ) negatively influenced the retention and efficacy of human MAPCs as well as IFN‐γ‐licensed MAPCs in the aGvHD model. PPARδ antagonism significantly enhanced the efficacy of human MAPCs when administered early in the humanized aGvHD model. COX‐2 expression in human MAPC was significantly decreased in IFN‐γ licensed MAPCs exposed to a PPARδ agonist. Importantly, MAPC exposure to the PPARδ antagonist in the presence of a COX‐2 inhibitor indomethacin before administration significantly reduced the efficacy of PPARδ antagonized MAPCs in the aGvHD humanized mouse model. This is the first study to demonstrate the importance of PPARδ in human MAPC efficacy in vivo and highlights the importance of understanding the disease microenvironment in which cell‐based therapies are to be administered. In particular, the presence of PPARδ ligands may negatively influence MAPC or MSC therapeutic efficacy.
Collapse
Affiliation(s)
- Fiona Carty
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Hazel Dunbar
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Ian J Hawthorne
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | | | | | - Wouter Van't Hof
- Athersys, Inc, Cleveland, Ohio, USA.,Cleveland Cord Blood Center, Cleveland, Ohio, USA
| | - Karen English
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
38
|
Lohmann S, Pool MBF, Rozenberg KM, Keller AK, Moers C, Møldrup U, Møller BK, Lignell SJM, Krag S, Sierra-Parraga JM, Lo Faro ML, Hunter J, Hoogduijn MJ, Baan CC, Leuvenink HGD, Ploeg RJ, Eijken M, Jespersen B. Mesenchymal stromal cell treatment of donor kidneys during ex vivo normothermic machine perfusion: A porcine renal autotransplantation study. Am J Transplant 2021; 21:2348-2359. [PMID: 33382194 DOI: 10.1111/ajt.16473] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/25/2023]
Abstract
Normothermic machine perfusion (NMP) of injured kidneys offers the opportunity for interventions to metabolically active organs prior to transplantation. Mesenchymal stromal cells (MSCs) can exert regenerative and anti-inflammatory effects in ischemia-reperfusion injury. The aims of this study were to evaluate the safety and feasibility of MSC treatment of kidneys during NMP using a porcine autotransplantation model, and examine potential MSC treatment-associated kidney improvements up to 14 days posttransplant. After 75 min of kidney warm ischemia, four experimental groups of n = 7 underwent 14 h of oxygenated hypothermic machine perfusion. In three groups this was followed by 240 min of NMP with infusion of vehicle, 10 million porcine, or 10 million human adipose-derived MSCs. All kidneys were autotransplanted after contralateral nephrectomy. MSC treatment did not affect perfusion hemodynamics during NMP or cause adverse effects at reperfusion, with 100% animal survival. MSCs did not affect plasma creatinine, glomerular filtration rate, neutrophil gelatinase-associated lipocalin concentrations or kidney damage assessed by histology during the 14 days, and MSCs retention was demonstrated in renal cortex. Infusing MSCs during ex vivo NMP of porcine kidneys was safe and feasible. Within the short posttransplant follow-up period, no beneficial effects of ex vivo MSC therapy could be demonstrated.
Collapse
Affiliation(s)
- Stine Lohmann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Merel B F Pool
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Anna K Keller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ulla Møldrup
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Stina J M Lignell
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Krag
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesus M Sierra-Parraga
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Maria L Lo Faro
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - James Hunter
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Martin J 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
| | - Henri G D Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rutger J Ploeg
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Immunology, 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
|
39
|
Marinescu CI, Preda MB, Burlacu A. A procedure for in vitro evaluation of the immunosuppressive effect of mouse mesenchymal stem cells on activated T cell proliferation. Stem Cell Res Ther 2021; 12:319. [PMID: 34090507 PMCID: PMC8178850 DOI: 10.1186/s13287-021-02344-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/19/2021] [Indexed: 02/08/2023] Open
Abstract
Background Mesenchymal stem/stromal cells (MSC) represent adult cells with multipotent capacity. Besides their capacity to differentiate into multiple lineages in vitro and in vivo, increasing evidence points towards the immunomodulatory capacity of these cells, as an important feature for their therapeutic power. Although not included in the minimal criteria established by the International Society for Cellular Therapy as a defining MSC attribute, demonstration of the immunomodulatory capacity of MSC can be useful for the characterization of these cells before being considered MSC. Methods Here we present a simple and reliable protocol by which the immunosuppressive effect of mouse bone marrow-derived MSC can be evaluated in vitro. It is based on the measuring of the proliferation of activated T cells cultured in direct contact with irradiated MSC. Results Our results showed that mouse MSC have a dose-dependent inhibitory effect on activated T cell proliferation, which can be quantified as a percentage of maximum proliferation. Our data shows that batch-to-batch variability can be determined within one or multiple experiments, by extracting the area under curve of T cell proliferation plotted against the absolute number of MSC in co-culture. Conclusions The validation of the immunosupressive capacity of MSC could be added to the characterization of the cells before being used in various MSC-based approaches to treat immunological diseases. Our results showed that mouse MSC have a dose-dependent inhibitory effect on activated T cell proliferation. The immunosuppressive properties of MSC vary between batches, but not between different passages of the same batch.
Collapse
Affiliation(s)
- Catalina-Iolanda Marinescu
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 B.P. Hasdeu Street, 050568, Bucharest, Romania
| | - Mihai Bogdan Preda
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 B.P. Hasdeu Street, 050568, Bucharest, Romania
| | - Alexandrina Burlacu
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", 8 B.P. Hasdeu Street, 050568, Bucharest, Romania.
| |
Collapse
|
40
|
Varkouhi AK, Monteiro APT, Tsoporis JN, Mei SHJ, Stewart DJ, Dos Santos CC. Genetically Modified Mesenchymal Stromal/Stem Cells: Application in Critical Illness. Stem Cell Rev Rep 2021; 16:812-827. [PMID: 32671645 PMCID: PMC7363458 DOI: 10.1007/s12015-020-10000-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Critical illnesses including sepsis, acute respiratory distress syndromes, ischemic cardiovascular disorders and acute organ injuries are associated with high mortality, morbidity as well as significant health care system expenses. While these diverse conditions require different specific therapeutic approaches, mesenchymal stem/stromal cell (MSCs) are multipotent cells capable of self-renewal, tri-lineage differentiation with a broad range regenerative and immunomodulatory activities, making them attractive for the treatment of critical illness. The therapeutic effects of MSCs have been extensively investigated in several pre-clinical models of critical illness as well as in phase I and II clinical cell therapy trials with mixed results. Whilst these studies have demonstrated the therapeutic potential for MSC therapy in critical illness, optimization for clinical use is an ongoing challenge. MSCs can be readily genetically modified by application of different techniques and tools leading to overexpress or inhibit genes related to their immunomodulatory or regenerative functions. Here we will review recent approaches designed to enhance the therapeutic potential of MSCs with an emphasis on the technology used to generate genetically modified cells, target genes, target diseases and the implication of genetically modified MSCs in cell therapy for critical illness.
Collapse
Affiliation(s)
- Amir K Varkouhi
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology (NJIT), Newark, NJ, 07102, USA
| | - Ana Paula Teixeira Monteiro
- Keenan and Li Ka Shing Knowledge Institute, University Health Toronto - St. Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - James N Tsoporis
- Keenan and Li Ka Shing Knowledge Institute, University Health Toronto - St. Michael's Hospital, Toronto, Ontario, Canada
| | - Shirley H J Mei
- Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, ON, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, ON, Canada
| | - Claudia C Dos Santos
- Keenan and Li Ka Shing Knowledge Institute, University Health Toronto - St. Michael's Hospital, Toronto, Ontario, Canada. .,Interdepartmental Division of Critical Care, St. Michael's Hospital/University of Toronto, 30 Bond Street, Room 4-008, Toronto, ON, M5B 1WB, Canada.
| |
Collapse
|
41
|
Preda MB, Neculachi CA, Fenyo IM, Vacaru AM, Publik MA, Simionescu M, Burlacu A. Short lifespan of syngeneic transplanted MSC is a consequence of in vivo apoptosis and immune cell recruitment in mice. Cell Death Dis 2021; 12:566. [PMID: 34075029 PMCID: PMC8169682 DOI: 10.1038/s41419-021-03839-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Mesenchymal stromal cells (MSC) are attractive tools for cell-based therapy, yet the mechanisms underlying their migration and survival post-transplantation are unclear. Accumulating evidence indicates that MSC apoptosis modulates both innate and adaptive immune responses which impact on MSC therapeutic effects. Using a dual tracking system, namely the Luciferase expression and VivoTrack680 labelling, and in vivo optical imaging, we investigated the survival and migration of MSC transplanted by various routes (intravenous, subcutaneous, intrapancreatic and intrasplenic) in order to identify the best delivery approach that provides an accumulation of therapeutic cells to the injured pancreas in the non-obese diabetic (NOD) mouse. The results showed that transplanted MSC had limited migration capacity, irrespective of the administration route, and were short-lived with almost total disappearance at 7 days after transplantation. Within one day after transplantation, cells activated hypoxia signalling pathways, followed by Caspase 3-mediated apoptosis. These were subsequently followed by local recruitment of immune cells at the transplantation site, and the engulfment of apoptotic MSC by macrophages. Our results argue for a "hit and die" mechanism of transplanted MSC. Further investigations will elucidate the molecular crosstalk between the inoculated and the host-immune cells.
Collapse
Affiliation(s)
- Mihai Bogdan Preda
- grid.418333.e0000 0004 1937 1389Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Carmen Alexandra Neculachi
- grid.418333.e0000 0004 1937 1389Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Ioana Madalina Fenyo
- grid.418333.e0000 0004 1937 1389Laboratory of Gene Regulation and Molecular Therapies, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Ana-Maria Vacaru
- grid.418333.e0000 0004 1937 1389Laboratory of Gene Regulation and Molecular Therapies, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Mihai Alin Publik
- grid.418333.e0000 0004 1937 1389Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Maya Simionescu
- grid.418333.e0000 0004 1937 1389Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania ,grid.418333.e0000 0004 1937 1389Laboratory of Gene Regulation and Molecular Therapies, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| | - Alexandrina Burlacu
- grid.418333.e0000 0004 1937 1389Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, Bucharest, Romania
| |
Collapse
|
42
|
Luo Y, Guo J, Zhang P, Cheuk YC, Jiang Y, Wang J, Xu S, Rong R. Mesenchymal Stem Cell Protects Injured Renal Tubular Epithelial Cells by Regulating mTOR-Mediated Th17/Treg Axis. Front Immunol 2021; 12:684197. [PMID: 34122446 PMCID: PMC8194268 DOI: 10.3389/fimmu.2021.684197] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
The increase in T helper 17 cell (Th17)-mediated pro-inflammatory response and decrease in regulatory T cell (Treg)-mediated anti-inflammatory effect aggravate renal tubular epithelial cell (RTEC) injury. However, increasing evidence indicated that mesenchymal stem cell (MSC) possessed the ability to control the imbalance between Th17 and Treg. Given that Th17 and Treg are derived from a common CD4+ T cell precursor, we summarize the current knowledge of MSC-mediated inhibition of the mammalian target of rapamycin (mTOR), which is a master regulator of CD4+ T cell polarization. During CD4+ T cell differentiation, mTOR signaling mediates Th17 and Treg differentiation via hypoxia-inducible factor-1α (HIF-1α)-dependent metabolic regulation and signaling pathway, as well as mTOR-mediated phosphorylation of signal transducer and activator of transcription (STAT) 3 and 5. Through interfering with mTOR signaling, MSC restrains CD4+ T cell differentiation into Th17, but in turn promotes Treg generation. Thus, this review indicates that MSC-mediated Th17-to-Treg polarization is expected to act as new immunotherapy for kidney injury.
Collapse
Affiliation(s)
- Yongsheng Luo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jingjing Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Pingbao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yamei Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jiyan Wang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Shanghai Medical College, Fudan University, Shanghai, China
| | - Shihao Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
43
|
Mesenchymal stromal cell therapeutic potency is dependent upon viability, route of delivery, and immune match. Blood Adv 2021; 4:1987-1997. [PMID: 32384543 DOI: 10.1182/bloodadvances.2020001711] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/12/2020] [Indexed: 12/11/2022] Open
Abstract
Culture-adapted bone marrow mesenchymal stromal cells (MSCs) deploy paracrine anti-inflammatory and tissue regenerative functionalities that can be harnessed as a living cell pharmaceutical product. Independent of clinical indication, a near majority of human clinical trials administer MSC IV, often with an allogeneic MSC cell product immediately after thawing from cryostorage. Despite hundreds of studies in a wide assortment of inflammatory, degenerative, and acute tissue injury syndromes, human clinical outcomes often fail to mirror promising rigorously conducted preclinical animal studies. Using a mouse model of toxic colitis, we demonstrate that replication fit MSCs harvested in log phase of growth have substantial impact on colitis clinical and pathologic endpoints when delivered subcutaneously or intraperitoneally, whereas the maximum tolerated IV bolus dosing failed to do so. We also demonstrate that heat-inactivated MSCs lose all therapeutic utility and the observation is mirrored by use of viable MSC administered immediately postthaw from cryostorage. Using luciferase transgenic MSC as donor cells, we demonstrate that transient in vivo engraftment is severely compromised when MSCs are dead or thawed and further demonstrate that MSC redosing is feasible in relapsing colitis, but only syngeneic MSCs lead to sustained improvement of clinical endpoints. These data support the notion that pharmaceutical potency of MSC requires viability and functional fitness. Reciprocally, IV administration of thawed MSC products may be biased against positive clinical outcomes for treatment of colitis and that extravascular administration of syngeneic, fit MSCs allows for effect in a recurrent therapy model.
Collapse
|
44
|
Mesenchymal stromal cells in hematopoietic cell transplantation. Blood Adv 2021; 4:5877-5887. [PMID: 33232479 DOI: 10.1182/bloodadvances.2020002646] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are widely recognized to possess potent immunomodulatory activity, as well as to stimulate repair and regeneration of diseased or damaged tissue. These fundamental properties suggest important applications in hematopoietic cell transplantation. Although the mechanisms of therapeutic activity in vivo are yet to be fully elucidated, MSCs seem to suppress lymphocytes by paracrine mechanisms, including secreted mediators and metabolic modulators. Most recently, host macrophage engulfment of apoptotic MSCs has emerged as an important contributor to the immune suppressive microenvironment. Although bone marrow-derived MSCs are the most commonly studied, the tissue source of MSCs may be a critical determinant of immunomodulatory function. The key application of MSC therapy in hematopoietic cell transplantation is to prevent or treat graft-versus-host disease (GVHD). The pathogenesis of GVHD reveals multiple potential targets. Moreover, the recently proposed concept of tissue tolerance suggests a new possible mechanism of MSC therapy for GVHD. Beyond GVHD, MSCs may facilitate hematopoietic stem cell engraftment, which could gain greater importance with increasing use of haploidentical transplantation. Despite many challenges and much doubt, commercial MSC products for pediatric steroid-refractory GVHD have been licensed in Japan, conditionally licensed in Canada and New Zealand, and have been recommended for approval by an FDA Advisory Committee in the United States. Here, we review key historical data in the context of the most salient recent findings to present the current state of MSCs as adjunct cell therapy in hematopoietic cell transplantation.
Collapse
|
45
|
Kowal JM, Möller S, Ali D, Figeac F, Barington T, Schmal H, Kassem M. Identification of a clinical signature predictive of differentiation fate of human bone marrow stromal cells. Stem Cell Res Ther 2021; 12:265. [PMID: 33941262 PMCID: PMC8091554 DOI: 10.1186/s13287-021-02338-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/19/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Transplantation of human bone marrow stromal cells (hBMSCs) is a promising therapy for bone regeneration due to their ability to differentiate into bone forming osteoblastic cells. However, transplanted hBMSCs exhibit variable capacity for bone formation resulting in inconsistent clinical outcome. The aim of the study was to identify a set of donor- and cell-related characteristics that detect hBMSCs with optimal osteoblastic differentiation capacity. METHODS We collected hBMSCs from 58 patients undergoing surgery for bone fracture. Clinical profile of the donors and in vitro characteristics of cultured hBMSCs were included in uni- and multivariable analysis to determine their predictive value for osteoblastic versus adipocytic differentiation capacity assessed by quantification of mineralized matrix and mature adipocyte formation, respectively. RESULTS We identified a signature that explained > 50% of variation in osteoblastic differentiation outcome which included the following positive predictors: donor sex (male), absence of osteoporosis diagnosis, intake of vitamin D supplements, higher fraction of CD146+, and alkaline phosphate (ALP+) cells. With the exception of vitamin D and ALP+ cells, these variables were also negative predictors of adipocytic differentiation. CONCLUSIONS Using a combination of clinical and cellular criteria, it is possible to predict differentiation outcome of hBMSCs. This signature may be helpful in selecting donor cells in clinical trials of bone regeneration.
Collapse
Affiliation(s)
- Justyna Magdalena Kowal
- Department of Endocrinology, Odense University Hospital, Odense, Denmark. .,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Sören Möller
- OPEN - Open Patient data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dalia Ali
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Florence Figeac
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Torben Barington
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hagen Schmal
- Department of Orthopedics and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Moustapha Kassem
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark
| |
Collapse
|
46
|
Galipeau J, Krampera M, Leblanc K, Nolta JA, Phinney DG, Shi Y, Tarte K, Viswanathan S, Martin I. Mesenchymal stromal cell variables influencing clinical potency: the impact of viability, fitness, route of administration and host predisposition. Cytotherapy 2021; 23:368-372. [PMID: 33714704 PMCID: PMC11708105 DOI: 10.1016/j.jcyt.2020.11.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/14/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
The International Society for Cell & Gene Therapy mesenchymal stromal cell (MSC) committee has been an interested observer of community interests in all matters related to MSC identity, mechanism of action, potency assessment and etymology, and it has regularly contributed to this conversation through a series of MSC pre-conferences and committee publications dealing with these matters. Arising from these reflections, the authors propose that an overlooked and potentially disruptive perspective is the impact of in vivo persistence on potency that is not predicted by surrogate cellular potency assays performed in vitro and how this translates to in vivo outcomes. Systemic delivery or extravascular implantation at sites removed from the affected organ system seems to be adequate in affecting clinical outcomes in many pre-clinical murine models of acute tissue injury and inflammatory pathology, including the recent European Medicines Agency-approved use of MSCs in Crohn-related fistular disease. The authors further propose that MSC viability and metabolic fitness likely dominate as a potency quality attribute, especially in recipients poised for salutary benefits as defined by emerging predictive biomarkers of response.
Collapse
Affiliation(s)
- Jacques Galipeau
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA.
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Katarina Leblanc
- Department of Laboratory Medicine, Center for Allogeneic Stem Cell Transplantation, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jan A Nolta
- Stem Cell Program, University of California Davis, Sacramento, California, USA
| | - Donald G Phinney
- Department of Molecular Therapeutics, Scripps Research Institute, Jupiter, Florida, USA
| | - Yufang Shi
- Institute for Translational Medicine, Soochow University, Suzhou, China
| | - Karin Tarte
- Établissement Français du Sang Bretagne, Institute for Health and Medical Research, University of Rennes, Rennes, France
| | - Sowmya Viswanathan
- Department of Medicine and Institute of Biomedical Engineering, Krembil Research Institute, University Health Network, University of Toronto, Toronto, Canada
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
47
|
Affiliation(s)
- Zachary W Wagoner
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA. .,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA. .,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA. .,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA. .,Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
48
|
Merino A, Sablik M, Korevaar SS, López-Iglesias C, Ortiz-Virumbrales M, Baan CC, Lombardo E, Hoogduijn MJ. Membrane Particles Derived From Adipose Tissue Mesenchymal Stromal Cells Improve Endothelial Cell Barrier Integrity. Front Immunol 2021; 12:650522. [PMID: 33897698 PMCID: PMC8058477 DOI: 10.3389/fimmu.2021.650522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/19/2021] [Indexed: 11/23/2022] Open
Abstract
Proinflammatory stimuli lead to endothelial injury, which results in pathologies such as cardiovascular diseases, autoimmune diseases, and contributes to alloimmune responses after organ transplantation. Both mesenchymal stromal cells (MSC) and the extracellular vesicles (EV) released by them are widely studied as regenerative therapy for the endothelium. However, for therapeutic application, the manipulation of living MSC and large-scale production of EV are major challenges. Membrane particles (MP) generated from MSC may be an alternative to the use of whole MSC or EV. MP are nanovesicles artificially generated from the membranes of MSC and possess some of the therapeutic properties of MSC. In the present study we investigated whether MP conserve the beneficial MSC effects on endothelial cell repair processes under inflammatory conditions. MP were generated by hypotonic shock and extrusion of MSC membranes. The average size of MP was 120 nm, and they showed a spherical shape. The effects of two ratios of MP (50,000; 100,000 MP per target cell) on human umbilical vein endothelial cells (HUVEC) were tested in a model of inflammation induced by TNFα. Confocal microscopy and flow cytometry showed that within 24 hours >90% of HUVEC had taken up MP. Moreover, MP ended up in the lysosomes of the HUVEC. In a co-culture system of monocytes and TNFα activated HUVEC, MP did not affect monocyte adherence to HUVEC, but reduced the transmigration of monocytes across the endothelial layer from 138 ± 61 monocytes per microscopic field in TNFα activated HUVEC to 61 ± 45 monocytes. TNFα stimulation induced a 2-fold increase in the permeability of the HUVEC monolayer measured by the translocation of FITC-dextran to the lower compartment of a transwell system. At a dose of 1:100,000 MP significantly decreased endothelial permeability (1.5-fold) respect to TNFα Stimulated HUVEC. Finally, MP enhanced the angiogenic potential of HUVEC in an in vitro Matrigel assay by stimulating the formation of angiogenic structures, such as percentage of covered area, total tube length, total branching points, total loops. In conclusion, MP show regenerative effects on endothelial cells, opening a new avenue for treatment of vascular diseases where inflammatory processes damage the endothelium.
Collapse
Affiliation(s)
- Ana Merino
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marta Sablik
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Sander S Korevaar
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute, FHML Maastricht University, Maastricht, Netherlands
| | | | - Carla C Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
49
|
García-Bernal D, García-Arranz M, Yáñez RM, Hervás-Salcedo R, Cortés A, Fernández-García M, Hernando-Rodríguez M, Quintana-Bustamante Ó, Bueren JA, García-Olmo D, Moraleda JM, Segovia JC, Zapata AG. The Current Status of Mesenchymal Stromal Cells: Controversies, Unresolved Issues and Some Promising Solutions to Improve Their Therapeutic Efficacy. Front Cell Dev Biol 2021; 9:650664. [PMID: 33796536 PMCID: PMC8007911 DOI: 10.3389/fcell.2021.650664] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) currently constitute the most frequently used cell type in advanced therapies with different purposes, most of which are related with inflammatory processes. Although the therapeutic efficacy of these cells has been clearly demonstrated in different disease animal models and in numerous human phase I/II clinical trials, only very few phase III trials using MSCs have demonstrated the expected potential therapeutic benefit. On the other hand, diverse controversial issues on the biology and clinical applications of MSCs, including their specific phenotype, the requirement of an inflammatory environment to induce immunosuppression, the relevance of the cell dose and their administration schedule, the cell delivery route (intravascular/systemic vs. local cell delivery), and the selected cell product (i.e., use of autologous vs. allogeneic MSCs, freshly cultured vs. frozen and thawed MSCs, MSCs vs. MSC-derived extracellular vesicles, etc.) persist. In the current review article, we have addressed these issues with special emphasis in the new approaches to improve the properties and functional capabilities of MSCs after distinct cell bioengineering strategies.
Collapse
Affiliation(s)
- David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Medicine Department, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, University of Murcia, Murcia, Spain.,Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain
| | - Mariano García-Arranz
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain
| | - Rosa M Yáñez
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Rosario Hervás-Salcedo
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Alfonso Cortés
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - María Fernández-García
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Miriam Hernando-Rodríguez
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Óscar Quintana-Bustamante
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Juan A Bueren
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Damián García-Olmo
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain
| | - Jose M Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Medicine Department, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, University of Murcia, Murcia, Spain.,Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain
| | - José C Segovia
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, Autonomous University of Madrid (UAM)), Madrid, Spain.,Centre for Cytometry and Fluorescence Microscopy, Complutense University, Madrid, Spain
| | - Agustín G Zapata
- Spanish Network of Cell Therapy (TerCel), Instituto de Salud Carlos III, Madrid, Spain.,Department of Cell Biology, Complutense University, Madrid, Spain
| |
Collapse
|
50
|
Mankuzhy PD, Ramesh ST, Thirupathi Y, Mohandas PS, Chandra V, Sharma TG. The preclinical and clinical implications of fetal adnexa derived mesenchymal stromal cells in wound healing therapy. Wound Repair Regen 2021; 29:347-369. [PMID: 33721373 DOI: 10.1111/wrr.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/06/2020] [Accepted: 03/01/2021] [Indexed: 11/28/2022]
Abstract
Mesenchymal stromal cells (MSCs) isolated from fetal adnexa namely amniotic membrane/epithelium, amniotic fluid and umbilical cord have hogged the limelight in recent times, as a proposed alternative to MSCs from conventional sources. These cells which are identified as being in a developmentally primitive state have many advantages, the most important being the non-invasive nature of their isolation procedures, absence of ethical concerns, proliferation potential, differentiation abilities and low immunogenicity. In the present review, we are focusing on the potential preclinical and clinical applications of different cell types of fetal adnexa, in wound healing therapy. We also discuss the isolation-culture methods, cell surface marker expression, multi-lineage differentiation abilities, immune-modulatory capabilities and their homing property. Different mechanisms involved in the wound healing process and the role of stromal cells in therapeutic wound healing are highlighted. Further, we summarize the findings of the cell delivery systems in skin lesion models and paracrine functions of their secretome in the wound healing process. Overall, this holistic review outlines the research findings of fetal adnexa derived MSCs, their usefulness in wound healing therapy in human as well as in veterinary medicine.
Collapse
Affiliation(s)
- Pratheesh D Mankuzhy
- Department of Physiology, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala, India
| | - Sreekumar T Ramesh
- Department of Physiology, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala, India
| | - Yasotha Thirupathi
- Physiology & Climatology Division, ICAR-Indian Veterinary Research Institute (Deemed University), Izatnagar, Uttar Pradesh, India
| | - Ponny S Mohandas
- Consultant Gynecologist, Department of Gynecology and Obstetrics, Meditrina Hospital, Ayathil, Kollam, Kerala, India
| | - Vikash Chandra
- Physiology & Climatology Division, ICAR-Indian Veterinary Research Institute (Deemed University), Izatnagar, Uttar Pradesh, India
| | - Taru Guttula Sharma
- Physiology & Climatology Division, ICAR-Indian Veterinary Research Institute (Deemed University), Izatnagar, Uttar Pradesh, India
| |
Collapse
|