1
|
Jacques V, Benaouadi S, Descamps JG, Reina N, Espagnolle N, Marsal D, Sainte-Marie Y, Boudet A, Pinto C, Farge T, Savagner F. Metabolic conditioning enhances human bmMSC therapy of doxorubicin-induced heart failure. Stem Cells 2024; 42:874-888. [PMID: 39133028 DOI: 10.1093/stmcls/sxae050] [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: 02/20/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic potential of bone marrow mesenchymal stromal cells (bmMSCs) to address heart failure needs improvement for better engraftment and survival. This study explores the role of metabolic sorting for human bmMSCs in coculture in vitro and on doxorubicin-induced heart failure mice models. Using functional, epigenetic, and gene expression approaches on cells sorted for mitochondrial membrane potential in terms of their metabolic status, we demonstrated that bmMSCs selected for their glycolytic metabolism presented proliferative advantage and resistance to oxidative stress thereby favoring cell engraftment. Therapeutic use of glycolytic bmMSCs rescued left ventricular ejection fraction and decreased fibrosis in mice models of acute heart failure. Metabolic changes were also related to epigenetic histone modifications such as lysine methylation. By targeting LSD1 (lysine-specific demethylase 1) as a conditioning agent to enhance the metabolic profile of bmMSCs, we deciphered the interplay between glycolysis and bmMSC functionality. Our study elucidates novel strategies for optimizing bmMSC-based treatments for heart failure, highlighting the metabolic properties of bmMSCs as a promising target for more effective cardiovascular regenerative therapies.
Collapse
Affiliation(s)
- Virginie Jacques
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
- Biochemistry Laboratory, IFB, CHU, 31059 Toulouse, France
| | - Sabrina Benaouadi
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
| | | | - Nicolas Reina
- Department of Orthopedic Surgery, Hôpital Pierre-Paul-Riquet, CHU de Toulouse, 31059 Toulouse, Cedex 9, France
- AMIS Laboratory-Laboratoire Anthropologie Moléculaire et Imagerie de Synthèse, Université de Toulouse, UMR 5288 CNRS, UPS, 31000 Toulouse, France
| | - Nicolas Espagnolle
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1031, France
| | | | - Yannis Sainte-Marie
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
| | - Alexandre Boudet
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
| | - Carla Pinto
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
| | - Thomas Farge
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
- Biochemistry Laboratory, IFB, CHU, 31059 Toulouse, France
| | - Frédérique Savagner
- Université Paul Sabatier, 31062 Toulouse, France
- Inserm UMR 1297 (Team 9), I2MC, 31400 Toulouse, France
- Biochemistry Laboratory, IFB, CHU, 31059 Toulouse, France
| |
Collapse
|
2
|
D’Angeli F, Granata G, Romano IR, Distefano A, Lo Furno D, Spila A, Leo M, Miele C, Ramadan D, Ferroni P, Li Volti G, Accardo P, Geraci C, Guadagni F, Genovese C. Biocompatible Poly(ε-Caprolactone) Nanocapsules Enhance the Bioavailability, Antibacterial, and Immunomodulatory Activities of Curcumin. Int J Mol Sci 2024; 25:10692. [PMID: 39409022 PMCID: PMC11476408 DOI: 10.3390/ijms251910692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Curcumin (Cur), the primary curcuminoid found in Curcuma longa L., has garnered significant attention for its potential anti-inflammatory and antibacterial properties. However, its hydrophobic nature significantly limits its bioavailability. Additionally, adipose-derived stem cells (ADSCs) possess immunomodulatory properties, making them useful for treating inflammatory and autoimmune conditions. This study aims to verify the efficacy of poly(ε-caprolactone) nanocapsules (NCs) in improving Cur's bioavailability, antibacterial, and immunomodulatory activities. The Cur-loaded nanocapsules (Cur-NCs) were characterized for their physicochemical properties (particle size, polydispersity index, Zeta potential, and encapsulation efficiency) and stability over time. A digestion test simulated the behavior of Cur-NCs in the gastrointestinal tract. Micellar phase analyses evaluated the Cur-NCs' bioaccessibility. The antibacterial activity of free Cur, NCs, and Cur-NCs against various Gram-positive and Gram-negative strains was determined using the microdilution method. ADSC viability, treated with Cur-NCs and Cur-NCs in the presence or absence of lipopolysaccharide, was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide assay. Additionally, ADSC survival was assessed through the Muse apoptotic assay. The expression of both pro-inflammatory (interleukin-1β and tumor necrosis factor-α) and anti-inflammatory (IL-10 and transforming growth factor-β) cytokines on ADSCs was evaluated by real-time polymerase chain reaction. The results demonstrated high stability post-gastric digestion of Cur-NCs and elevated bioaccessibility of Cur post-intestinal digestion. Moreover, Cur-NCs exhibited antibacterial activity against Escherichia coli without affecting Lactobacillus growth. No significant changes in the viability and survival of ADSCs were observed under the experimental conditions. Finally, Cur-NCs modulated the expression of both pro- and anti-inflammatory cytokines in ADSCs exposed to inflammatory stimuli. Collectively, these findings highlight the potential of Cur-NCs to enhance Cur's bioavailability and therapeutic efficacy, particularly in cell-based treatments for inflammatory diseases and intestinal dysbiosis.
Collapse
Affiliation(s)
- Floriana D’Angeli
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Giuseppe Granata
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Antonella Spila
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Mariantonietta Leo
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Chiara Miele
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Dania Ramadan
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Patrizia Ferroni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Paolo Accardo
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Corrada Geraci
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Fiorella Guadagni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Carlo Genovese
- Department of Medicine and Surgery, “Kore” University of Enna, Contrada Santa Panasia, 94100 Enna, Italy;
- Nacture S.r.l, Spin-Off University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
3
|
Gordon J, Borlongan CV. An update on stem cell therapy for stroke patients: Where are we now? J Cereb Blood Flow Metab 2024; 44:1469-1479. [PMID: 38639015 PMCID: PMC11418600 DOI: 10.1177/0271678x241227022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 04/20/2024]
Abstract
With a foundation built upon initial work from the 1980s demonstrating graft viability in cerebral ischemia, stem cell transplantation has shown immense promise in promoting survival, enhancing neuroprotection and inducing neuroregeneration, while mitigating both histological and behavioral deficits that frequently accompany ischemic stroke. These findings have led to a number of clinical trials that have thoroughly supported a strong safety profile for stem cell therapy in patients but have generated variable efficacy. As preclinical evidence continues to expand through the investigation of new cell lines and optimization of stem cell delivery, it remains critical for translational models to adhere to the protocols established through basic scientific research. With the recent shift in approach towards utilization of stem cells as a conjunctive therapy alongside standard thrombolytic treatments, key issues including timing, route of administration, and stem cell type must each be appropriately translated from the laboratory in order to resolve the question of stem cell efficacy for cerebral ischemia that ultimately will enhance therapeutics for stroke patients towards improving quality of life.
Collapse
Affiliation(s)
- Jonah Gordon
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
4
|
Arki MK, Moeinabadi-Bidgoli K, Niknam B, Mohammadi P, Hassan M, Hossein-Khannazer N, Vosough M. Immunomodulatory performance of GMP-compliant, clinical-grade mesenchymal stromal cells from four different sources. Heliyon 2024; 10:e24948. [PMID: 38312681 PMCID: PMC10835001 DOI: 10.1016/j.heliyon.2024.e24948] [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: 09/09/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/06/2024] Open
Abstract
Inflammatory and autoimmune diseases are among the most challenging disorders for health care professionals that require systemic immune suppression which associates with various side effects. Mesenchymal stromal cells (MSCs) are capable of regulating immune responses, mainly through paracrine effects and cell-cell contact. Since MSCs are advanced therapy medicinal products (ATMPs), they must follow Good Manufacturing Practice (GMP) regulations to ensure their safety and efficacy. In this study, we evaluated the immunomodulatory effects of GMP-compliant clinical grade MSCs obtained from four different sources (bone marrow, adipose tissue, Wharton's Jelly, and decidua tissue) on allogeneic peripheral blood mononuclear cells (PBMCs). Our results revealed that WJ-MSCs were the most successful group in inhibiting PBMC proliferation as confirmed by BrdU analysis. Moreover, WJ-MSCs were the strongest group in enhancing the regulatory T cell population of PBMCs. WJ-MSCs also had the highest secretory profile of prostaglandin E2 (PGE-2), anti-inflammatory cytokine, while interleukin-10 (IL-10) secretion was highest in the DS-MSC group. DS-MSCs also had the lowest secretion of IL-12 and IL-17 inflammatory cytokines. Transcriptome analysis revealed that WJ-MSCs had the lowest expression of IL-6, while DS-MSCs were the most potent group in the expression of immunomodulatory factors such as hepatocyte growth factor (HGF) and transforming growth factor-β (TGF- β). Taken together, our results indicated that GMP-compliant Wharton's Jelly and decidua-derived MSCs showed the best immunomodulatory performance considering paracrine factors.
Collapse
Affiliation(s)
- Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Moeinabadi-Bidgoli
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Niknam
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvaneh Mohammadi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83, Stockholm, Sweden
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83, Stockholm, Sweden
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
5
|
Kuçi Z, Piede N, Vogelsang K, Pfeffermann LM, Wehner S, Salzmann-Manrique E, Stais M, Kreyenberg H, Bonig H, Bader P, Kuçi S. Expression of HLA-DR by mesenchymal stromal cells in the platelet lysate era: an obsolete release criterion for MSCs? J Transl Med 2024; 22:39. [PMID: 38195462 PMCID: PMC10775607 DOI: 10.1186/s12967-023-04684-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND According to the definition of the International Society for Cell and Gene Therapy (ISCT), mesenchymal stromal cells (MSCs) do not express HLA-DR. This phenotypic marker as a release criterion for clinical use was established at a time when MSCs were expanded in fetal bovine serum (FBS)-containing media. Replacement of FBS with platelet lysate (PLs) as a medium supplement induced a significantly higher fraction of MSCs to express MHC class II antigens. METHODS As this raised concerns that such MSCs may play the role of antigen-presenting cells for T cells, in the current study, we studied major factors that may induce HLA-DR on MSCs by means of flow cytometry and real-time polymerase chain reaction. The immunomodulatory potential of MSCs was assessed by a mixed lymphocyte reaction. RESULTS Our results demonstrated that a very low percentage of generated and expanded MSCs in FBS express HLA-DR (median: 1.1%, range: 0.3-22%) compared to MSCs generated and expanded in PLs (median: 28.4%, range: 3.3-73.7%). Analysis of the cytokine composition of ten PLs showed a significant positive correlation between the levels of IL-1β, IL-4, IL-10, IL-17, bFGF and expression of HLA-DR, in contrast to no correlation with the age of MSC donors and HLA-DR (r = 0.21). Both MSCs expressing low and high levels of HLA-DR expressed class II transactivator (CIITA), a master gene coding for these molecules. Our results demonstrate for the first time that MSCs with constitutively high levels of HLA-DR also express moderate levels of indoleamine 2,3-dioxygenase (IDO). Treatment of MSCs with multiple doses of TGF-β1 at passage 0 (P0) and passage 1 (P1) completely abrogated HLA-DR and IDO expression. In contrast, treatment of MSCs with a single dose of TGF-β1 after P0 only partially reduced the expression of HLA-DR and CIITA. Remarkably, increased expression of HLA-DR on MSCs that constitutively express high levels of this antigen after overnight incubation with IFN-γ was rather unaffected by incubation with TGF-β1. However, treatment of MSCs with TGF-β1 for 24 h completely abrogated constitutive expression of IDO. CONCLUSIONS Irrespective of HLA-DR expression at the population level, all MSC preparations significantly inhibited the proliferation of stimulated peripheral blood mononuclear cells, indicating that HLA-DR represents an obsolete release marker for the clinical use of MSCs.
Collapse
Affiliation(s)
- Zyrafete Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Natascha Piede
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Kathrin Vogelsang
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Lisa-Marie Pfeffermann
- Institute for Transfusion Medicine and Immunohematology, Goethe University and German Red Cross Blood Service BaWüHe, Institute Frankfurt, Frankfurt, Germany
| | - Sibylle Wehner
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Miriam Stais
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Hermann Kreyenberg
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University and German Red Cross Blood Service BaWüHe, Institute Frankfurt, Frankfurt, Germany
| | - Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Selim Kuçi
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
6
|
Mahmoud M, Abdel-Rasheed M, Galal ER, El-Awady RR. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev Rep 2024; 20:175-205. [PMID: 37962697 PMCID: PMC10799834 DOI: 10.1007/s12015-023-10654-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Human adipose tissue-derived stem/stromal cells (hASCs) are adult multipotent mesenchymal stem/stromal cells with immunomodulatory capacities. Here, we present up-to-date knowledge on the impact of different experimental and donor-related factors on hASC immunoregulatory functions in vitro. The experimental determinants include the immunological status of hASCs relative to target immune cells, contact vs. contactless interaction, and oxygen tension. Factors such as the ratio of hASCs to immune cells, the cellular context, the immune cell activation status, and coculture duration are also discussed. Conditioning of hASCs with different approaches before interaction with immune cells, hASC culture in xenogenic or xenofree culture medium, hASC culture in two-dimension vs. three-dimension with biomaterials, and the hASC passage number are among the experimental parameters that greatly may impact the hASC immunosuppressive potential in vitro, thus, they are also considered. Moreover, the influence of donor-related characteristics such as age, sex, and health status on hASC immunomodulation in vitro is reviewed. By analysis of the literature studies, most of the indicated determinants have been investigated in broad non-standardized ranges, so the results are not univocal. Clear conclusions cannot be drawn for the fine-tuned scenarios of many important factors to set a standard hASC immunopotency assay. Such variability needs to be carefully considered in further standardized research. Importantly, field experts' opinions may help to make it clearer.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| | - Eman Reda Galal
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Rehab R El-Awady
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
7
|
Annamalai C, Kute V, Sheridan C, Halawa A. Hematopoietic cell-based and non-hematopoietic cell-based strategies for immune tolerance induction in living-donor renal transplantation: A systematic review. Transplant Rev (Orlando) 2023; 37:100792. [PMID: 37709652 DOI: 10.1016/j.trre.2023.100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/24/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Despite its use to prevent acute rejection, lifelong immunosuppression can adversely impact long-term patient and graft outcomes. In theory, immunosuppression withdrawal is the ultimate goal of kidney transplantation, and is made possible by the induction of immunological tolerance. The purpose of this paper is to review the safety and efficacy of immune tolerance induction strategies in living-donor kidney transplantation, both chimerism-based and non-chimerism-based. The impact of these strategies on transplant outcomes, including acute rejection, allograft function and survival, cost, and immune monitoring, will also be discussed. MATERIALS AND METHODS Databases such as PubMed, Scopus, and Web of Science, as well as additional online resources such as EBSCO, were exhaustively searched. Adult living-donor kidney transplant recipients who developed chimerism-based tolerance after concurrent bone marrow or hematopoietic stem cell transplantation or those who received non-chimerism-based, non-hematopoietic cell therapy using mesenchymal stromal cells, dendritic cells, or regulatory T cells were studied between 2000 and 2021. Individual sources of evidence were evaluated critically, and the strength of evidence and risk of bias for each outcome of the transplant tolerance study were assessed. RESULTS From 28,173 citations, 245 studies were retrieved after suitable exclusion and duplicate removal. Of these, 22 studies (2 RCTs, 11 cohort studies, 6 case-control studies, and 3 case reports) explicitly related to both interventions (chimerism- and non-chimerism-based immune tolerance) were used in the final review process and were critically appraised. According to the findings, chimerism-based strategies fostered immunotolerance, allowing for the safe withdrawal of immunosuppressive medications. Cell-based therapy, on the other hand, frequently did not induce tolerance except for minimising immunosuppression. As a result, the rejection rates, renal allograft function, and survival rates could not be directly compared between these two groups. While chimerism-based tolerance protocols posed safety concerns due to myelosuppression, including infections and graft-versus-host disease, cell-based strategies lacked these adverse effects and were largely safe. There was a lack of direct comparisons between HLA-identical and HLA-disparate recipients, and the cost implications were not examined in several of the retrieved studies. Most studies reported successful immunosuppressive weaning lasting at least 3 years (ranging up to 11.4 years in some studies), particularly with chimerism-based therapy, while only a few investigators used immune surveillance techniques. The studies reviewed were often limited by selection, classification, ascertainment, performance, and attrition bias. CONCLUSIONS This review demonstrates that chimerism-based hematopoietic strategies induce immune tolerance, and a substantial number of patients are successfully weaned off immunosuppression. Despite the risk of complications associated with myelosuppression. Non-chimerism-based, non-hematopoietic cell protocols, on the other hand, have been proven to facilitate immunosuppression minimization but seldom elicit immunological tolerance. However, the results of this review must be interpreted with caution because of the non-randomised study design, potential confounding, and small sample size of the included studies. Further validation and refinement of tolerogenic protocols in accordance with local practice preferences is also warranted, with an emphasis on patient selection, cost ramifications, and immunological surveillance based on reliable tolerance assays.
Collapse
Affiliation(s)
- Chandrashekar Annamalai
- Postgraduate School of Medicine, Institute of Teaching and Learning, Faculty of Health and Life Sciences, University of Liverpool, UK.
| | - Vivek Kute
- Nephrology and Transplantation, Institute of Kidney Diseases and Research Center and Dr. H L Trivedi Institute of Transplantation Sciences (IKDRC-ITS), Ahmedabad, India
| | - Carl Sheridan
- Department of Eye and Vision Science, Ocular Cell Transplantation, Faculty of Health and Life Sciences, University of Liverpool, UK
| | - Ahmed Halawa
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
8
|
Ding L, Oh S, Shrestha J, Lam A, Wang Y, Radfar P, Warkiani ME. Scaling up stem cell production: harnessing the potential of microfluidic devices. Biotechnol Adv 2023; 69:108271. [PMID: 37844769 DOI: 10.1016/j.biotechadv.2023.108271] [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: 04/29/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Stem cells are specialised cells characterised by their unique ability to both self-renew and transform into a wide array of specialised cell types. The widespread interest in stem cells for regenerative medicine and cultivated meat has led to a significant demand for these cells in both research and practical applications. Despite the growing need for stem cell manufacturing, the industry faces significant obstacles, including high costs for equipment and maintenance, complicated operation, and low product quality and yield. Microfluidic technology presents a promising solution to the abovementioned challenges. As an innovative approach for manipulating liquids and cells within microchannels, microfluidics offers a plethora of advantages at an industrial scale. These benefits encompass low setup costs, ease of operation and multiplexing, minimal energy consumption, and the added advantage of being labour-free. This review presents a thorough examination of the prominent microfluidic technologies employed in stem cell research and explores their promising applications in the burgeoning stem cell industry. It thoroughly examines how microfluidics can enhance cell harvesting from tissue samples, facilitate mixing and cryopreservation, streamline microcarrier production, and efficiently conduct cell separation, purification, washing, and final cell formulation post-culture.
Collapse
Affiliation(s)
- Lin Ding
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia.
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Alan Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yaqing Wang
- School of Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Payar Radfar
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia..
| |
Collapse
|
9
|
Fang YH, Wang SPH, Liao IC, Tsai KJ, Huang PH, Yang PJ, Yen CJ, Liu PY, Shan YS, Liu YW. HLA-E high /HLA-G high /HLA-II low Human iPSC-Derived Cardiomyocytes Exhibit Low Immunogenicity for Heart Regeneration. Adv Healthc Mater 2023; 12:e2301186. [PMID: 37672681 DOI: 10.1002/adhm.202301186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/31/2023] [Indexed: 09/08/2023]
Abstract
Although human pluripotent stem cells (hPSCs)-derived cardiomyocytes (hPSC-CMs) can remuscularize infarcted hearts and restore post-infarct cardiac function, post-transplant rejection resulting from human leukocyte antigen (HLA) mismatching is an enormous obstacle. It is crucial to identify hypoimmunogenic hPSCs for allogeneic cell therapy. This study is conducted to demonstrate the immune privilege of HLA-Ehigh /HLA-Ghigh /HLA-IIlow human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs). Ischemia-reperfusion surgery is done to create transmural myocardial infarction in rats. At post-infarct 4 days, hPSC-CMs (1.0×107 cells per kg), including human embryonic stem cell-derived cardiomyocytes (hESC-CMs), HLA-Elow/HLA-Glow/HLA-IIhigh hiPSC-CMs, and HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs, are injected into the infarcted myocardium. Under the treatment of very low dose cyclosporine A (CsA), only HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs survive in vivo and improved post-infarct cardiac function with infarct size reduction. HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs activate the SHP-1 signaling pathway of natural killer (NK) cells and cytotoxic T cells to evade attack by NK cells and cytotoxic T cells. Herein, it is demonstrated that using a clinically relevant CsA dose, HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSC-CMs repair the infarcted myocardium and restore the post-infarct heart function. HLA-Ehigh /HLA-Ghigh /HLA-IIlow hiPSCs are less immunogenic and may serve as platforms for regeneration medicine.
Collapse
Affiliation(s)
- Yi-Hsien Fang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Saprina P H Wang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - I-Chuang Liao
- Department of Pathology, Chi-Mei Medical Center, Tainan, 71004, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Po-Hsien Huang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Pei-Jung Yang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Chia-Jui Yen
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Yen-Wen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| |
Collapse
|
10
|
Mahmoud M, Abdel-Rasheed M. Influence of type 2 diabetes and obesity on adipose mesenchymal stem/stromal cell immunoregulation. Cell Tissue Res 2023; 394:33-53. [PMID: 37462786 PMCID: PMC10558386 DOI: 10.1007/s00441-023-03801-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/21/2023] [Indexed: 10/07/2023]
Abstract
Type 2 diabetes (T2D), associated with obesity, represents a state of metabolic inflammation and oxidative stress leading to insulin resistance and progressive insulin deficiency. Adipose-derived stem cells (ASCs) are adult mesenchymal stem/stromal cells identified within the stromal vascular fraction of adipose tissue. These cells can regulate the immune system and possess anti-inflammatory properties. ASCs are a potential therapeutic modality for inflammatory diseases including T2D. Patient-derived (autologous) rather than allogeneic ASCs may be a relatively safer approach in clinical perspectives, to avoid occasional anti-donor immune responses. However, patient characteristics such as body mass index (BMI), inflammatory status, and disease duration and severity may limit the therapeutic utility of ASCs. The current review presents human ASC (hASC) immunoregulatory mechanisms with special emphasis on those related to T lymphocytes, hASC implications in T2D treatment, and the impact of T2D and obesity on hASC immunoregulatory potential. hASCs can modulate the proliferation, activation, and functions of diverse innate and adaptive immune cells via direct cell-to-cell contact and secretion of paracrine mediators and extracellular vesicles. Preclinical studies recommend the therapeutic potential of hASCs to improve inflammation and metabolic indices in a high-fat diet (HFD)-induced T2D disease model. Discordant data have been reported to unravel intact or detrimentally affected immunomodulatory functions of ASCs, isolated from patients with obesity and/or T2D patients, in vitro and in vivo. Numerous preconditioning strategies have been introduced to potentiate hASC immunomodulation; they are also discussed here as possible options to potentiate the immunoregulatory functions of hASCs isolated from patients with obesity and T2D.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| |
Collapse
|
11
|
Shekari F, Alibhai FJ, Baharvand H, Börger V, Bruno S, Davies O, Giebel B, Gimona M, Salekdeh GH, Martin‐Jaular L, Mathivanan S, Nelissen I, Nolte‐’t Hoen E, O'Driscoll L, Perut F, Pluchino S, Pocsfalvi G, Salomon C, Soekmadji C, Staubach S, Torrecilhas AC, Shelke GV, Tertel T, Zhu D, Théry C, Witwer K, Nieuwland R. Cell culture-derived extracellular vesicles: Considerations for reporting cell culturing parameters. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e115. [PMID: 38939735 PMCID: PMC11080896 DOI: 10.1002/jex2.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/18/2023] [Accepted: 09/17/2023] [Indexed: 06/29/2024]
Abstract
Cell culture-conditioned medium (CCM) is a valuable source of extracellular vesicles (EVs) for basic scientific, therapeutic and diagnostic applications. Cell culturing parameters affect the biochemical composition, release and possibly the function of CCM-derived EVs (CCM-EV). The CCM-EV task force of the Rigor and Standardization Subcommittee of the International Society for Extracellular Vesicles aims to identify relevant cell culturing parameters, describe their effects based on current knowledge, recommend reporting parameters and identify outstanding questions. While some recommendations are valid for all cell types, cell-specific recommendations may need to be established for non-mammalian sources, such as bacteria, yeast and plant cells. Current progress towards these goals is summarized in this perspective paper, along with a checklist to facilitate transparent reporting of cell culturing parameters to improve the reproducibility of CCM-EV research.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP‐TDC), Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | | | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in BiologyUniversity of Science and CultureTehranIran
| | - Verena Börger
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Stefania Bruno
- Department of Medical Sciences and Molecular Biotechnology CenterUniversity of TorinoTurinItaly
| | - Owen Davies
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Mario Gimona
- GMP UnitSpinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI‐TReCS) and Research Program “Nanovesicular Therapies” Paracelsus Medical UniversitySalzburgAustria
| | | | - Lorena Martin‐Jaular
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular ScienceLa Trobe UniversityMelbourneVICAustralia
| | - Inge Nelissen
- VITO (Flemish Institute for Technological Research), Health departmentBoeretangBelgium
| | - Esther Nolte‐’t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | - Francesca Perut
- Biomedical Science and Technologies and Nanobiotechnology LabIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Gabriella Pocsfalvi
- Institute of Biosciences and BioResourcesNational Research CouncilNaplesItaly
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
| | - Carolina Soekmadji
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | | | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)SPBrazil
| | - Ganesh Vilas Shelke
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
| | - Dandan Zhu
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVICAustralia
| | - Clotilde Théry
- Institut Curie, INSERM U932 and Curie CoreTech Extracellular VesiclesPSL Research UniversityParisFrance
| | - Kenneth Witwer
- Departments of Molecular and Comparative Pathobiology and Neurology and Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, Amsterdam University Medical CentersLocation AMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
12
|
Wang Z, Liao Y, Wang C, Tang C, Fang C, Luo J, Liu H, Mo X, Wang Z, Shen L, Wang J, Chen X, Yin Z, Li J, Shen W. Stem cell-based therapeutic strategies for rotator cuff tendinopathy. J Orthop Translat 2023; 42:73-81. [PMID: 37664079 PMCID: PMC10470406 DOI: 10.1016/j.jot.2023.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
Rotator cuff tendinopathy is a common musculoskeletal disorder that imposes significant health and economic burden. Stem cell therapy has brought hope for tendon healing in patients with final stage rotator cuff tendinopathy. Some clinical trials have confirmed the effectiveness of stem cell therapy for rotator cuff tendinopathy, but its application has not been promoted and approved. There are still many issues that should be solved prior to using stem cell therapy in clinical applications. The optimal source and dose of stem cells for rotator cuff tendinopathy should be determined. We also proposed novel prospective approaches that can overcome cell population heterogeneity and standardize patient types for stem cell applications. The translational potential of this article This review explores the optimal sources of stem cells for rotator cuff tendinopathy and the principles for selecting stem cell dosages. Key strategies are provided for stem cell population standardization and recipient selection.
Collapse
Affiliation(s)
- Zetao Wang
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Youguo Liao
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Cailian Fang
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Junchao Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianan Mo
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zicheng Wang
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Lingfang Shen
- Air Force Health Care Center for Special Services, Hangzhou, China
| | | | - Xiao Chen
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zi Yin
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianyou Li
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Weiliang Shen
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Loisel S, Lansiaux P, Rossille D, Ménard C, Dulong J, Monvoisin C, Bescher N, Bézier I, Latour M, Cras A, Farge D, Tarte K. Regulatory B Cells Contribute to the Clinical Response After Bone Marrow-Derived Mesenchymal Stromal Cell Infusion in Patients With Systemic Sclerosis. Stem Cells Transl Med 2023; 12:194-206. [PMID: 36928395 PMCID: PMC10108721 DOI: 10.1093/stcltm/szad010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/09/2023] [Indexed: 03/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have recently emerged as an interesting therapeutic approach for patients with progressive systemic sclerosis (SSc), a rare and life-threatening orphan autoimmune disease. Whereas MSC immunomodulatory potential is considered as a central mechanism for their clinical benefit, very few data are available on the impact of MSCs on immune cell subsets in vivo. In the current extended study of a phase I/II clinical trial exploring the injection of a single dose of allogeneic bone marrow-MSCs (alloBM-MSCs) in patients with severe SSc (NCT02213705), we performed a longitudinal in-depth characterization of circulating immune cells in 19 MSC-treated patients, including 14 responders and 5 non-responders. By a combination of flow cytometry and transcriptomic analyses, we highlighted an increase in circulating CD24hiCD27posCD38lo/neg memory B cells, the main IL-10-producing regulatory B cell (Breg) subset, and an upregulation of IL10 expression in ex-vivo purified B cells, specifically in responder patients, early after the alloBM-MSC infusion. In addition, a deeper alteration of the B-cell compartment before alloBM-MSC treatment, including a higher expression of profibrotic cytokines IL6 and TGFβ by sorted B cells was associated with a non-responder clinical status. Finally, BM-MSCs were able to directly upregulate IL-10 production in activated B cells in vitro. These data suggest that cytokine-producing B cells, in particular Breg, are pivotal effectors of BM-MSC therapeutic activity in SSc. Their quantification as activity biomarkers in MSC potency assays and patient selection criteria may be considered to reach optimal clinical benefit when designing MSC-based clinical trials.
Collapse
Affiliation(s)
- Séverine Loisel
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04), CRMR Maladies auto-immunes et thérapie cellulaire (MATHEC), Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, Hôpital St-Louis, Paris, France
- Université de Paris Cité, IRSL, Recherche clinique appliquée à l’hématologie, URP 3518, Paris, France
| | - Delphine Rossille
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Cédric Ménard
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Joëlle Dulong
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Céline Monvoisin
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Nadège Bescher
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Isabelle Bézier
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Maëlle Latour
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Audrey Cras
- Cell Therapy Unit, Saint Louis Hospital, Assistance-Publique Hôpitaux de Paris, Paris, France
- UMR1140, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France
| | - Dominique Farge
- Unité de Médecine Interne (UF 04), CRMR Maladies auto-immunes et thérapie cellulaire (MATHEC), Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, Hôpital St-Louis, Paris, France
- Université de Paris Cité, IRSL, Recherche clinique appliquée à l’hématologie, URP 3518, Paris, France
- Department of Medicine, McGill University, Montreal, Canada
| | - Karin Tarte
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
14
|
Secretome of Mesenchymal Stromal Cells as a Possible Innovative Therapeutic Tool in Facial Nerve Injury Treatment. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8427200. [PMID: 36691473 PMCID: PMC9867597 DOI: 10.1155/2023/8427200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Facial nerve palsy is a serious neurological condition that strongly affects patient everyday life. Standard treatments provide insufficient improvement and are burdened with the risk of severe complications, e.g., facial synkinesis. Mesenchymal stromal cell-based therapies are a novel and extensively developed field which offers new treatment approaches with promising results in regards to the nervous tissue regeneration. The potential of mesenchymal stromal cells (MSCs) to aid the regeneration of damaged nerves has been demonstrated in several preclinical models, as well as in several clinical trials. However, therapies based on cell transplantation are difficult to standardize in the manner similar to that of routine clinical practices. On the other hand, treatments based on mesenchymal stromal cell secretome harness the proregenerative features of mesenchymal stromal cells but relay on a product with measurable parameters that can be put through standardization procedures and deliver a fully controllable end-product. Utilization of mesenchymal stromal cell secretome allows the controlled dosage and standardization of the components to maximize the therapeutic potential and ensure safety of the end-product.
Collapse
|
15
|
Miclau K, Hambright WS, Huard J, Stoddart MJ, Bahney CS. Cellular expansion of MSCs: Shifting the regenerative potential. Aging Cell 2023; 22:e13759. [PMID: 36536521 PMCID: PMC9835588 DOI: 10.1111/acel.13759] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/14/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal-derived stromal or progenitor cells, commonly called "MSCs," have attracted significant clinical interest for their remarkable abilities to promote tissue regeneration and reduce inflammation. Recent studies have shown that MSCs' therapeutic effects, originally attributed to the cells' direct differentiation capacity into the tissue of interest, are largely driven by the biomolecules the cells secrete, including cytokines, chemokines, growth factors, and extracellular vesicles containing miRNA. This secretome coordinates upregulation of endogenous repair and immunomodulation in the local microenvironment through crosstalk of MSCs with host tissue cells. Therapeutic applications for MSCs and their secretome-derived products often involve in vitro monolayer expansion. However, consecutive passaging of MSCs significantly alters their therapeutic potential, inducing a broad shift from a pro-regenerative to a pro-inflammatory phenotype. A consistent by-product of in vitro expansion of MSCs is the onset of replicative senescence, a state of cell arrest characterized by an increased release of proinflammatory cytokines and growth factors. However, little is known about changes in the secretome profile at different stages of in vitro expansion. Some culture conditions and bioprocessing techniques have shown promise in more effectively retaining the pro-regenerative and anti-inflammatory MSC phenotype throughout expansion. Understanding how in vitro expansion conditions influence the nature and function of MSCs, and their associated secretome, may provide key insights into the underlying mechanisms driving these alterations. Elucidating the dynamic and diverse changes in the MSC secretome at each stage of in vitro expansion is a critical next step in the development of standardized, safe, and effective MSC-based therapies.
Collapse
Affiliation(s)
- Katherine Miclau
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
- Orthopaedic Trauma Institute (OTI)University of California San FranciscoSan FranciscoCaliforniaUSA
| | - William S. Hambright
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
| | - Martin J. Stoddart
- Orthopaedic Trauma Institute (OTI)University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Chelsea S. Bahney
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
- AO Research Institute DavosDavosSwitzerland
| |
Collapse
|
16
|
Porter AP, Pirlot BM, Dyer K, Uwazie CC, Nguyen J, Turner C, Rajan D, Hematti P, Chinnadurai R. Conglomeration of T- and B-Cell Matrix Responses Determines the Potency of Human Bone Marrow Mesenchymal Stromal Cells. Stem Cells 2022; 40:1134-1148. [PMID: 36056823 DOI: 10.1093/stmcls/sxac064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023]
Abstract
Cell manufacturing facilities need to define the potency of mesenchymal stromal cells (MSCs) as cellular therapeutics in advanced clinical trials or marketing approval. Since MSCs' mechanism of action in humans is not well defined, more than a single functional property of MSCs needs to be captured as a surrogate measure of potency utilizing assay matrix technologies. However, the current limitation is the sole investigation of MSC-mediated T-cell suppression as a surrogate measure of potency. We investigated the effect of MSCs on B-cell matrix responses to be incorporated into the assay matrix potency analytical system. Our results demonstrate that MSCs inhibit B-cell differentiation and block pan-antibody secretion upon activation of B cells in the PBMCs. In contrast, MSCs are inferior in blocking B-cell matrix responses when purified B cells are used. Mechanistic analysis has demonstrated that MSC-mediated inhibition of B-cell matrix responses is non-contact dependent and Tryptophan metabolic pathway plays a major role, akin to the mechanism of MSC-mediated T-cell suppression. MSCs also inhibit both T-cell and B-cell responses when both of these lymphoid populations are concurrently activated in the PBMCs. Secretome analysis of MSC and T/B cell-activated PBMC cocultures identified direct and inverse correlative matrix signatures between humoral antibody isotypes and secretory molecules. The current analysis of the combined and concomitant investigation of T-cell and B-cell matrix responses fulfills the potency assay matrix strategy by incorporating MSCs' interaction with more than a single inflammatory immune responder.
Collapse
Affiliation(s)
- Amanda P Porter
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Bonnie M Pirlot
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Kalyn Dyer
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Crystal C Uwazie
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Jimmy Nguyen
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Caitlin Turner
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Devi Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin Madison, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| |
Collapse
|
17
|
da Graça Cabreira M, Wang X, Critsinelis A, Setegne M, Lotfi P, Wan YW, Barrios G, Mei Z, Gee AP, Buja LM, Perin E. Environmental oxygen affects ex vivo growth and proliferation of mesenchymal progenitors by modulating mitogen-activated protein kinase and mammalian target of rapamycin signaling. Cytotherapy 2022; 24:1201-1210. [PMID: 36109320 DOI: 10.1016/j.jcyt.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/16/2022] [Accepted: 06/13/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Stem and progenitor cells of hematopoietic and mesenchymal lineages reside in the bone marrow under low oxygen (O2) saturation. O2 levels used in ex vivo expansion of multipotent mesenchymal stromal cells (MSCs) affect proliferation, metabolism and differentiation. METHODS Using cell-based assays and transcriptome and proteome data, the authors compared MSC cultures simultaneously grown under a conventional 19.95% O2 atmosphere or at 5% O2. RESULTS In 5% O2, MSCs showed better proliferation and higher self-renewal ability, most probably sustained by enhanced signaling activity of mitogen-activated protein kinase and mammalian target of rapamycin pathways. Non-oxidative glycolysis-based energy metabolism supported growth and proliferation in 5% O2 cultures, whereas MSCs grown under 19.95% O2 also utilized oxidative phosphorylation. Cytoprotection mechanisms used by cells under 5% O2 differed from 19.95% O2 suggesting differences in the triggers of cell stress between these two O2 conditions. CONCLUSIONS Based on the potential benefits for the growth and metabolism of MSCs, the authors propose the use of 5% O2 for MSC culture.
Collapse
Affiliation(s)
| | - Xiaohong Wang
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mekedlawit Setegne
- Chemistry-Biology Interface Predoctoral Training Program, Stanford University, Stanford, California, USA
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Gabriela Barrios
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, USA
| | - Zhuyong Mei
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Adrian P Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Louis Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| | - Emerson Perin
- Center for Clinical Research, Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
18
|
Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and Mesenchymal Stem Cells: Basic Concepts, Challenges and Strategies. BIOLOGY 2022; 11:1678. [PMID: 36421393 PMCID: PMC9687158 DOI: 10.3390/biology11111678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023]
Abstract
Aging and frailty are complex processes implicating multifactorial mechanisms, such as replicative senescence, oxidative stress, mitochondrial dysfunction, or autophagy disorder. All of these mechanisms drive dramatic changes in the tissue environment, such as senescence-associated secretory phenotype factors and inflamm-aging. Thus, there is a demand for new therapeutic strategies against the devastating effects of the aging and associated diseases. Mesenchymal stem cells (MSC) participate in a "galaxy" of tissue signals (proliferative, anti-inflammatory, and antioxidative stress, and proangiogenic, antitumor, antifibrotic, and antimicrobial effects) contributing to tissue homeostasis. However, MSC are also not immune to aging. Three strategies based on MSC have been proposed: remove, rejuvenate, or replace the senescent MSC. These strategies include the use of senolytic drugs, antioxidant agents and genetic engineering, or transplantation of younger MSC. Nevertheless, these strategies may have the drawback of the adverse effects of prolonged use of the different drugs used or, where appropriate, those of cell therapy. In this review, we propose the new strategy of "Exogenous Restitution of Intercellular Signalling of Stem Cells" (ERISSC). This concept is based on the potential use of secretome from MSC, which are composed of molecules such as growth factors, cytokines, and extracellular vesicles and have the same biological effects as their parent cells. To face this cell-free regenerative therapy challenge, we have to clarify key strategy aspects, such as establishing tools that allow us a more precise diagnosis of aging frailty in order to identify the therapeutic requirements adapted to each case, identify the ideal type of MSC in the context of the functional heterogeneity of these cellular populations, to optimize the mass production and standardization of the primary materials (cells) and their secretome-derived products, to establish the appropriate methods to validate the anti-aging effects and to determine the most appropriate route of administration for each case.
Collapse
Affiliation(s)
- Maria Fraile
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Arancha Martín
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Emergency, Hospital Universitario de Cabueñes, Los Prados, 395, 33394 Gijon, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Surgery, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| |
Collapse
|
19
|
Fuentes P, Torres MJ, Arancibia R, Aulestia F, Vergara M, Carrión F, Osses N, Altamirano C. Dynamic Culture of Mesenchymal Stromal/Stem Cell Spheroids and Secretion of Paracrine Factors. Front Bioeng Biotechnol 2022; 10:916229. [PMID: 36046670 PMCID: PMC9421039 DOI: 10.3389/fbioe.2022.916229] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, conditioned medium (CM) obtained from the culture of mesenchymal stromal/stem cells (MSCs) has been shown to effectively promote tissue repair and modulate the immune response in vitro and in different animal models, with potential for application in regenerative medicine. Using CM offers multiple advantages over the implantation of MSCs themselves: 1) simpler storage, transport, and preservation requirements, 2) avoidance of the inherent risks of cell transplantation, and 3) potential application as a ready-to-go biologic product. For these reasons, a large amount of MSCs research has focused on the characterization of the obtained CM, including soluble trophic factors and vesicles, preconditioning strategies for enhancing paracrine secretion, such as hypoxia, a three-dimensional (3D) environment, and biochemical stimuli, and potential clinical applications. In vitro preconditioning strategies can increase the viability, proliferation, and paracrine properties of MSCs and therefore improve the therapeutic potential of the cells and their derived products. Specifically, dynamic cultivation conditions, such as fluid flow and 3D aggregate culture, substantially impact cellular behaviour. Increased levels of growth factors and cytokines were observed in 3D cultures of MSC grown on orbital or rotatory shaking platforms, in stirred systems, such as spinner flasks or stirred tank reactors, and in microgravity bioreactors. However, only a few studies have established dynamic culture conditions and protocols for 3D aggregate cultivation of MSCs as a scalable and reproducible strategy for CM production. This review summarizes significant advances into the upstream processing, mainly the dynamic generation and cultivation of MSC aggregates, for de CM manufacture and focuses on the standardization of the soluble factor production.
Collapse
Affiliation(s)
- Paloma Fuentes
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María José Torres
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Rodrigo Arancibia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Francisco Aulestia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Mauricio Vergara
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Flavio Carrión
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Nelson Osses
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- CREAS, Centro Regional de Estudios en Alimentos Saludables, Valparaíso, Chile
- *Correspondence: Claudia Altamirano,
| |
Collapse
|
20
|
Eiro N, Fraile M, González-Jubete A, González LO, Vizoso FJ. Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges. Int J Mol Sci 2022; 23:ijms23168905. [PMID: 36012170 PMCID: PMC9408403 DOI: 10.3390/ijms23168905] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are an example of chronic diseases affecting 40% of the population, which involved tissue damage and an inflammatory process not satisfactorily controlled with current therapies. Data suggest that mesenchymal stem cells (MSC) may be a therapeutic option for these processes, and especially for IBD, due to their multifactorial approaches such as anti-inflammatory, anti-oxidative stress, anti-apoptotic, anti-fibrotic, regenerative, angiogenic, anti-tumor, or anti-microbial. However, MSC therapy is associated with important limitations as safety issues, handling difficulties for therapeutic purposes, and high economic cost. MSC-derived secretome products (conditioned medium or extracellular vesicles) are therefore a therapeutic option in IBD as they exhibit similar effects to their parent cells and avoid the issues of cell therapy. In this review, we proposed further studies to choose the ideal tissue source of MSC to treat IBD, the implementation of new standardized production strategies, quality controls and the integration of other technologies, such as hydrogels, which may improve the therapeutic effects of derived-MSC secretome products in IBD.
Collapse
Affiliation(s)
- Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| | - Maria Fraile
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | | | - Luis O. González
- Department of Anatomical Pathology, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Department of Surgery, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| |
Collapse
|
21
|
Shen Z, Wang D, Yu C, Peng Y, Cheng L, Zhang Y. Quantitative profiling of differentially expressed oxylipins in ADSCs under proinflammatory cytokines stimulation. Biomed Chromatogr 2022; 36:e5452. [PMID: 35853831 DOI: 10.1002/bmc.5452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/08/2022]
Abstract
MSCs have been proven to have anti-inflammatory capabilities, but the mechanisms are still under investigation. Recently, oxylipins have been identified to be related to the immuno-regulation function of MSCs, but the MSCs derived oxylipins, especially under the stimulation of versatile pro-inflammatory cytokines, have never been comprehensively analyzed. In the present research, an UPLC-MS/MS method was employed to identify and quantify the oxylipin profiles of ADSCs under cytokines stimulation (IL-1β, TNF-α, IFN-𝛾and TNF-α + IFN-𝛾). The differentially produced oxylipins between experimental groups wereanalyzed and compared. The elevated level of lipoxygenase-15 (LOX-15) mRNA was further verified by qRT-PCR analysis. From the targeted 71 oxylipins, we detected and quantified 57 oxylipins, while 14 were not detected. Distinctive from other cytokines, ADSCs activated by the combination of IFN-𝛾 and TNF-α up-produced LOX-15 products, 7-HDHA and 15-HEPE, which were metabolized from DHA and EPA respectively and involved in the pro-resolution phase of inflammation. The results reported here make a first step towards a comprehensive characterization of MSCs derived oxylipins under different proinflammatory cytokines stimulation. And the findings may lay a fundamental foundation for MSCs based therapies and further determine the ways to optimize the therapeutic potential of MSCs.
Collapse
Affiliation(s)
- Zhengze Shen
- Department ofPharmacy, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Wang
- Gastroenterology Department, Jiulongpo People's Hospital, Chongqing, China
| | - Chaoqun Yu
- College ofPharmacy, Chongqing Medical University, Chongqing, China
| | - Yongbo Peng
- College ofPharmacy, Chongqing Medical University, Chongqing, China
| | - Lifang Cheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Zhang
- Department ofPharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory ofMolecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Tamouza R, Volt F, Richard JR, Wu CL, Bouassida J, Boukouaci W, Lansiaux P, Cappelli B, Scigliuolo GM, Rafii H, Kenzey C, Mezouad E, Naamoune S, Chami L, Lejuste F, Farge D, Gluckman E. Possible Effect of the use of Mesenchymal Stromal Cells in the Treatment of Autism Spectrum Disorders: A Review. Front Cell Dev Biol 2022; 10:809686. [PMID: 35865626 PMCID: PMC9294632 DOI: 10.3389/fcell.2022.809686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Autism spectrum disorder (ASD) represents a set of heterogeneous neurodevelopmental conditions defined by impaired social interactions and repetitive behaviors. The number of reported cases has increased over the past decades, and ASD is now a major public health burden. So far, only treatments to alleviate symptoms are available, with still unmet need for an effective disease treatment to reduce ASD core symptoms. Genetic predisposition alone can only explain a small fraction of the ASD cases. It has been reported that environmental factors interacting with specific inter-individual genetic background may induce immune dysfunctions and contribute to the incidence of ASD. Such dysfunctions can be observed at the central level, with increased microglial cells and activation in ASD brains or in the peripheral blood, as reflected by high circulating levels of pro-inflammatory cytokines, abnormal activation of T-cell subsets, presence of auto-antibodies and of dysregulated microbiota profiles. Altogether, the dysfunction of immune processes may result from immunogenetically-determined inefficient immune responses against a given challenge followed by chronic inflammation and autoimmunity. In this context, immunomodulatory therapies might offer a valid therapeutic option. Mesenchymal stromal cells (MSC) immunoregulatory and immunosuppressive properties constitute a strong rationale for their use to improve ASD clinical symptoms. In vitro studies and pre-clinical models have shown that MSC can induce synapse formation and enhance synaptic function with consequent improvement of ASD-like symptoms in mice. In addition, two preliminary human trials based on the infusion of cord blood-derived MSC showed the safety and tolerability of the procedure in children with ASD and reported promising clinical improvement of core symptoms. We review herein the immune dysfunctions associated with ASD provided, the rationale for using MSC to treat patients with ASD and summarize the current available studies addressing this subject.
Collapse
Affiliation(s)
- Ryad Tamouza
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
- *Correspondence: Ryad Tamouza,
| | - Fernanda Volt
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Jean-Romain Richard
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Ching-Lien Wu
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Jihène Bouassida
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Wahid Boukouaci
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Barbara Cappelli
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Graziana Maria Scigliuolo
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Hanadi Rafii
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Chantal Kenzey
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Esma Mezouad
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Soumia Naamoune
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Leila Chami
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Florian Lejuste
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Dominique Farge
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Eliane Gluckman
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| |
Collapse
|
23
|
Mesenchymal stem cells: A living carrier for active tumor-targeted delivery. Adv Drug Deliv Rev 2022; 185:114300. [PMID: 35447165 DOI: 10.1016/j.addr.2022.114300] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022]
Abstract
The strategy of using mesenchymal stem cells (MSCs) as a living carrier for active delivery of therapeutic agents targeting tumor sites has been attempted in a wide range of studies to validate the feasibility and efficacy for tumor treatment. This approach reveals powerful tumor targeting and tumor penetration. In addition, MSCs have been confirmed to actively participate in immunomodulation of the tumor microenvironment. Thus, MSCs are not inert delivery vehicles but have a strong impact on the fate of tumor cells. In this review, these active properties of MSCs are addressed to highlight the advantages and challenges of using MSCs for tumor-targeted delivery. In addition, some of the latest examples of using MSCs to carry a variety of anti-tumor agents for tumor-targeted therapy are summarized. Recent technologies to improve the performance and safety of this delivery strategy will be introduced. The advances, applications, and challenges summarized in this review will provide a general understanding of this promising strategy for actively delivering drugs to tumor tissues.
Collapse
|
24
|
Palombella S, Perucca Orfei C, Castellini G, Gianola S, Lopa S, Mastrogiacomo M, Moretti M, de Girolamo L. Systematic review and meta-analysis on the use of human platelet lysate for mesenchymal stem cell cultures: comparison with fetal bovine serum and considerations on the production protocol. Stem Cell Res Ther 2022; 13:142. [PMID: 35379348 PMCID: PMC8981660 DOI: 10.1186/s13287-022-02815-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/10/2022] [Indexed: 11/24/2022] Open
Abstract
Mesenchymal stem cell (MSC) culturing for cell therapies needs a step forward to be routinely used in clinical settings. Main concerns regard the use of animal origin reagents, in particular supplementing the culture medium with FBS. Lately, Human Platelet Lysate (HPL) has been proposed as animal-free alternative, described as an excellent supplement for culturing MSCs. The aim of this systematic review was to analyze the current literature on the effect of HPL and FBS on ASCs and BMSCs. The primary outcome was the proliferation rate of cells cultured with FBS and HPL. Differences in terms of doubling time (DT) and population doubling (PD) were evaluated by meta-analysis, subgrouping data according to the cell type. A total of 35 articles were included. BMSCs and ASCs were used in 65.7% (23) and 28.6% (10) studies, respectively. Only two studies included both cell types. Overall, 22 studies were eligible for the meta-analysis. Among them, 9 articles described ASCs and 13 BMSCs. The results showed that BMSCs and ASCs cultured with 10% HPL and 5% HPL have lower DT and higher PD compared to cells cultured with 10% FBS. A possible correlation between the DT decrease and the application of at least 3 freeze/thaw cycles to induce platelet lysis was found. Additionally, HPL increased VEGF secretion and maintained the immuno-modulatory abilities for both cell types. The clarification reported here of the higher efficiency of HPL compared to FBS can help the transition of the scientific community towards clinical-related procedures.
Collapse
Affiliation(s)
- Silvia Palombella
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy
| | - Greta Castellini
- Unit of Clinical Epidemiology, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy
| | - Silvia Gianola
- Unit of Clinical Epidemiology, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy
| | | | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy.,Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, Laboratories for Translational Research (LRT), 6500, Bellinzona, Switzerland.,Department of Surgery, Ente Ospedaliero Cantonale, Service of Orthopaedics and Traumatology, 6962, Lugano, Switzerland.,Faculty of Biomedical Sciences, Euler Institute, USI, 6900, Lugano, Switzerland
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, 20161, Milan, Italy.
| |
Collapse
|
25
|
Tan KX, Chang T, Lin XL. Secretomes as an emerging class of bioactive ingredients for enhanced cosmeceutical applications. Exp Dermatol 2022; 31:674-688. [PMID: 35338666 DOI: 10.1111/exd.14570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/23/2022] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Abstract
Skin aging is predominantly caused by either intrinsic or extrinsic factors, leading to undesirable skin features. Advancements in both molecular and cellular fields have created possibilities in developing novel stem cell-derived active ingredients for cosmeceutical applications and the beauty industry. Mesenchymal stromal cell (MSC)-derived secretomes or conditioned media hold great promise for advancing skin repair and regeneration due to the presence of varying cytokines. These cytokines signal our cells and trigger biological mechanisms associated with anti-inflammatory, antioxidant, anti-aging, proliferative, and immunomodulatory effects. In this review, we discuss the potential of MSC secretomes as novel biomaterials for skincare and rejuvenation by illustrating their mechanism of action related to wound healing, anti-aging, and whitening properties. The advantages and disadvantages of secretomes are compared to both plant-based and animal-derived extracts. In addition, this paper reviews the current safety standards, regulations, market products and research work related to the cosmeceutical applications of secretomes along with strategies to maintain and improve the therapeutic efficacy and production of secretomes. The future outlook of beauty industry is also presented. Lastly, we highlight significant challenges to be addressed for the clinical realization of MSC secretomes-based skin therapies as well as providing perspectives for the future direction of secretomes.
Collapse
Affiliation(s)
- Kei-Xian Tan
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| | - Trixie Chang
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| | - Xiang-Liang Lin
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| |
Collapse
|
26
|
CD40L-expressing CD4+ T cells prime adipose-derived stromal cells to produce inflammatory chemokines. Cytotherapy 2022; 24:500-507. [DOI: 10.1016/j.jcyt.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/21/2021] [Accepted: 01/20/2022] [Indexed: 11/18/2022]
|
27
|
Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022; 24:456-472. [DOI: 10.1016/j.jcyt.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
|
28
|
Farge D, Loisel S, Resche-Rigon M, Lansiaux P, Colmegna I, Langlais D, Charles C, Pugnet G, Maria ATJ, Chatelus E, Martin T, Hachulla E, Kheav VD, Lambert NC, Wang H, Michonneau D, Martinaud C, Sensebé L, Cras A, Tarte K. Safety and preliminary efficacy of allogeneic bone marrow-derived multipotent mesenchymal stromal cells for systemic sclerosis: a single-centre, open-label, dose-escalation, proof-of-concept, phase 1/2 study. THE LANCET RHEUMATOLOGY 2022. [DOI: 10.1016/s2665-9913(21)00326-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
29
|
Mesenchymal Stem Cell-Based Therapy as a New Approach for the Treatment of Systemic Sclerosis. Clin Rev Allergy Immunol 2022; 64:284-320. [PMID: 35031958 DOI: 10.1007/s12016-021-08892-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis (SSc) is an intractable autoimmune disease with unmet medical needs. Conventional immunosuppressive therapies have modest efficacy and obvious side effects. Targeted therapies with small molecules and antibodies remain under investigation in small pilot studies. The major breakthrough was the development of autologous haematopoietic stem cell transplantation (AHSCT) to treat refractory SSc with rapidly progressive internal organ involvement. However, AHSCT is contraindicated in patients with advanced visceral involvement. Mesenchymal stem cells (MSCs) which are characterized by immunosuppressive, antifibrotic and proangiogenic capabilities may be a promising alternative option for the treatment of SSc. Multiple preclinical and clinical studies on the use of MSCs to treat SSc are underway. However, there are several unresolved limitations and safety concerns of MSC transplantation, such as immune rejections and risks of tumour formation, respectively. Since the major therapeutic potential of MSCs has been ascribed to their paracrine signalling, the use of MSC-derived extracellular vesicles (EVs)/secretomes/exosomes as a "cell-free" therapy might be an alternative option to circumvent the limitations of MSC-based therapies. In the present review, we overview the current knowledge regarding the therapeutic efficacy of MSCs in SSc, focusing on progresses reported in preclinical and clinical studies using MSCs, as well as challenges and future directions of MSC transplantation as a treatment option for patients with SSc.
Collapse
|
30
|
Mebarki M, Iglicki N, Marigny C, Abadie C, Nicolet C, Churlaud G, Maheux C, Boucher H, Monsel A, Menasché P, Larghero J, Faivre L, Cras A. Development of a human umbilical cord-derived mesenchymal stromal cell-based advanced therapy medicinal product to treat immune and/or inflammatory diseases. Stem Cell Res Ther 2021; 12:571. [PMID: 34774107 PMCID: PMC8590372 DOI: 10.1186/s13287-021-02637-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) revealed their key role in immune regulation, offering promising therapeutic perspectives for immune and inflammatory diseases. We aimed to develop a production process of an UC-MSC-based product and then to characterize UC-MSC properties and immunomodulatory activities in vitro, related to their clinical use and finally, to transfer this technology to a good manufacturing practice (GMP) compliant facility, to manufacture an advanced therapy medicinal product (ATMP). Methods Fifteen human umbilical cords (UCs) were collected to develop the production process. Three batches of UC-MSCs from a single donor were characterized at basal state and after in vitro pro-inflammatory stimulation by interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα). Proliferation, immunophenotype, activation markers’ expression and the inhibition of T cell proliferation were assessed. Finally, this technology was transferred to a GMP-compliant facility to manufacture an UC-MSC-based ATMP, from a single donor, using the explant method followed by the establishment of master and work cell stocks. Results Twelve UCs were processed successfully allowing to isolate UC-MSCs with doubling time and population doubling remaining stable until passage 4. CD90, CD105, CD73, CD44, CD29, CD166 expression was positive; CD14, CD45, CD31, HLA-DR, CD40, CD80 and CD86 expression was negative, while CD146 and HLA-ABC expression was heterogeneous. Cell morphology, proliferation and immunophenotype were not modified by inflammatory treatment. Indoleamine 2,3-dioxygenase (IDO) expression was significantly induced by IFNγ and IFNγ + TNFα versus non-treated cells. Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expression was induced significantly after priming. T cell proliferation was significantly decreased in the presence of UC-MSCs in a dose-dependent manner. This inhibitory effect was improved by IFNγ or IFNγ + TNFα, at UC-MSCs:PBMC ratio 1:10 and 1:30, whereas only IFNγ allowed to decrease significantly T cell proliferation at ratio 1:100. The manufacturing process of the UC-MSC-based ATMP was qualified and authorized by the French regulatory agency for clinical use (NCT04333368). Conclusion This work allowed to develop an investigational UC-MSC-based ATMP authorized for clinical use. Our results showed that an inflammatory environment preserves the biological properties of UC-MSCs with an improvement of their immunomodulatory functions. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02637-7.
Collapse
Affiliation(s)
- Miryam Mebarki
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France. .,INSERM U976, Université de Paris, 75010, Paris, France. .,Faculté de Pharmacie, Université de Paris, 75006, Paris, France.
| | | | - Céline Marigny
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Camille Abadie
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Claire Nicolet
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France
| | - Guillaume Churlaud
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Camille Maheux
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Hélène Boucher
- AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Antoine Monsel
- Unité de Soins Intensifs Et Département de Biothérapies, inflammation et immunopathologie, AP-HP, Hôpital La Pitié-Salpêtrière, 75013, Paris, France.,INSERM UMR-S 959, Université Sorbonne, 75012, Paris, France
| | - Philippe Menasché
- Département de Chirurgie Cardiovasculaire, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Jérôme Larghero
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France.,INSERM U976, Université de Paris, 75010, Paris, France.,AP-HP, Hôpital Saint-Louis, Centre MEARY de Thérapie Cellulaire Et Génique, 75010, Paris, France
| | - Lionel Faivre
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France.,INSERM U976, Université de Paris, 75010, Paris, France
| | - Audrey Cras
- INSERM Centre d'investigation Clinique de Biothérapies CBT501, AP-HP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, 75010, Paris, France. .,INSERM UMR1140, Université de Paris, 75006, Paris, France. .,Faculté de Pharmacie, Université de Paris, 75006, Paris, France.
| |
Collapse
|
31
|
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: 134] [Impact Index Per Article: 33.5] [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
|
32
|
Mesenchymal Stromal Cells: an Antimicrobial and Host-Directed Therapy for Complex Infectious Diseases. Clin Microbiol Rev 2021; 34:e0006421. [PMID: 34612662 DOI: 10.1128/cmr.00064-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is an urgent need for new antimicrobial strategies for treating complex infections and emerging pathogens. Human mesenchymal stromal cells (MSCs) are adult multipotent cells with antimicrobial properties, mediated through direct bactericidal activity and modulation of host innate and adaptive immune cells. More than 30 in vivo studies have reported on the use of human MSCs for the treatment of infectious diseases, with many more studies of animal MSCs in same-species models of infection. MSCs demonstrate potent antimicrobial effects against the major classes of human pathogens (bacteria, viruses, fungi, and parasites) across a wide range of infection models. Mechanistic studies have yielded important insight into their immunomodulatory and bactericidal activity, which can be enhanced through various forms of preconditioning. MSCs are being investigated in over 80 clinical trials for difficult-to-treat infectious diseases, including sepsis and pulmonary, intra-abdominal, cutaneous, and viral infections. Completed trials consistently report MSCs to be safe and well tolerated, with signals of efficacy against some infectious diseases. Although significant obstacles must be overcome to produce a standardized, affordable, clinical-grade cell therapy, these studies suggest that MSCs may have particular potential as an adjunct therapy in complex or resistant infections.
Collapse
|
33
|
Strategies to address mesenchymal stem/stromal cell heterogeneity in immunomodulatory profiles to improve cell-based therapies. Acta Biomater 2021; 133:114-125. [PMID: 33857693 DOI: 10.1016/j.actbio.2021.03.069] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/15/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) have gained immense attention over the past two decades due to their multipotent differentiation potential and pro-regenerative and immunomodulatory cytokine secretory profiles. Their ability to modulate the host immune system and promote tolerance has prompted several allogeneic and autologous hMSC-based clinical trials for the treatment of graft-versus-host disease and several other immune-induced disorders. However, clinical success beyond safety is still controversial and highly variable, with inconclusive therapeutic benefits and little mechanistic explanation. This clinical variability has been broadly attributed to inconsistent MSC sourcing, phenotypic characterization, variable potency, and non-standard isolation protocols, leading to functional heterogeneity among administered MSCs. Homogeneous MSC populations are proposed to yield more predictable, reliable biological responses and clinically meaningful properties relevant to cell-based therapies. Limited comparisons of heterogeneous MSCs with homogenous MSCs are reported. This review addresses this gap in the literature with a critical analysis of strategies aimed at decreasing MSC heterogeneity concerning their reported immunomodulatory profiles. STATEMENT OF SIGNIFICANCE: This review collates, summarizes, and critically analyzes published strategies that seek to improve homogeneity in immunomodulatory functioning MSC populations intended as cell therapies to treat immune-based disorders, such as graft-vs-host-disease. No such review for MSC therapies, immunomodulatory profiles and cell heterogeneity analysis is published. Since MSCs represent the most clinically studied experimental cell therapy platform globally for which there remains no US domestic marketing approval, insights into MSC challenges in therapeutic product development are imperative to providing solutions for immunomodulatory variabilities.
Collapse
|
34
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|
35
|
Importance of lymphocyte-stromal cell interactions in autoimmune and inflammatory rheumatic diseases. Nat Rev Rheumatol 2021; 17:550-564. [PMID: 34345021 DOI: 10.1038/s41584-021-00665-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
Interactions between lymphocytes and stromal cells have an important role in immune cell development and responses. During inflammation, stromal cells contribute to inflammation, from induction to chronicity or resolution, through direct cell interactions and through the secretion of pro-inflammatory and anti-inflammatory mediators. Stromal cells are imprinted with tissue-specific phenotypes and contribute to site-specific lymphocyte recruitment. During chronic inflammation, the modified pro-inflammatory microenvironment leads to changes in the stromal cells, which acquire a pathogenic phenotype. At the site of inflammation, infiltrating B cells and T cells interact with stromal cells. These interactions induce a plasma cell-like phenotype in B cells and T cells, associated with secretion of immunoglobulins and inflammatory cytokines, respectively. B cells and T cells also influence the stromal cells, inducing cell proliferation, molecular changes and cytokine production. This positive feedback loop contributes to disease chronicity. This Review describes the importance of these cell interactions in chronic inflammation, with a focus on human disease, using three selected autoimmune and inflammatory diseases: rheumatoid arthritis, psoriatic arthritis (and psoriasis) and systemic lupus erythematosus. Understanding the importance and disease specificity of these interactions could provide new therapeutic options.
Collapse
|
36
|
Rapid Production and Genetic Stability of Human Mesenchymal Progenitor Cells Derived from Human Somatic Cell Nuclear Transfer-Derived Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22179238. [PMID: 34502145 PMCID: PMC8431643 DOI: 10.3390/ijms22179238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Pluripotent stem cell-derived mesenchymal progenitor cells (PSC-MPCs) are primarily derived through two main methods: three-dimensional (3D) embryoid body-platform (EB formation) and the 2D direct differentiation method. We recently established somatic cell nuclear transfer (SCNT)-PSC lines and showed their stemness. In the present study, we produced SCNT-PSC-MPCs using a novel direct differentiation method, and the characteristics, gene expression, and genetic stability of these MPCs were compared with those derived through EB formation. The recovery and purification of SCNT-PSC-Direct-MPCs were significantly accelerated compared to those of the SCNT-PSC-EB-MPCs, but both types of MPCs expressed typical surface markers and exhibited similar proliferation and differentiation potentials. Additionally, the analysis of gene expression patterns using microarrays showed very similar patterns. Moreover, array CGH analysis showed that both SCNT-PSC-Direct-MPCs and SCNT-PSC-EB-MPCs exhibited no significant differences in copy number variation (CNV) or single-nucleotide polymorphism (SNP) frequency. These results indicate that SCNT-PSC-Direct-MPCs exhibited high genetic stability even after rapid differentiation into MPCs, and the rate at which directly derived MPCs reached a sufficient number was higher than that of MPCs derived through the EB method. Therefore, we suggest that the direct method of differentiating MPCs from SCNT-PSCs can improve the efficacy of SCNT-PSCs applied to allogeneic transplantation.
Collapse
|
37
|
Labedz-Maslowska A, Szkaradek A, Mierzwinski T, Madeja Z, Zuba-Surma E. Processing and Ex Vivo Expansion of Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells for the Development of an Advanced Therapy Medicinal Product for use in Humans. Cells 2021; 10:cells10081908. [PMID: 34440677 PMCID: PMC8392403 DOI: 10.3390/cells10081908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue (AT) represents a commonly used source of mesenchymal stem/stromal cells (MSCs) whose proregenerative potential has been widely investigated in multiple clinical trials worldwide. However, the standardization of the manufacturing process of MSC-based cell therapy medicinal products in compliance with the requirements of the local authorities is obligatory and will allow us to obtain the necessary permits for product administration according to its intended use. Within the research phase (RD), we optimized the protocols used for the processing and ex vivo expansion of AT-derived MSCs (AT-MSCs) for the development of an Advanced Therapy Medicinal Product (ATMP) for use in humans. Critical process parameters (including, e.g., the concentration of enzyme used for AT digestion, cell culture conditions) were identified and examined to ensure the high quality of the final product containing AT-MSCs. We confirmed the identity of isolated AT-MSCs as MSCs and their trilineage differentiation potential according to the International Society for Cellular Therapy (ISCT) recommendations. Based on the conducted experiments, in-process quality control (QC) parameters and acceptance criteria were defined for the manufacturing of hospital exemption ATMP (HE-ATMP). Finally, we conducted a validation of the manufacturing process in a GMP facility. In the current study, we presented a process approach leading to the optimization of processing and the ex vivo expansion of AT-MSCs for the development of ATMP for use in humans.
Collapse
Affiliation(s)
- Anna Labedz-Maslowska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (A.S.); (Z.M.)
| | - Agnieszka Szkaradek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (A.S.); (Z.M.)
- Cell & Tissue Culture Laboratory, Jagiellonian Center of Innovation in Krakow, 30-384 Krakow, Poland;
| | - Tomasz Mierzwinski
- Cell & Tissue Culture Laboratory, Jagiellonian Center of Innovation in Krakow, 30-384 Krakow, Poland;
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (A.S.); (Z.M.)
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (A.S.); (Z.M.)
- Correspondence: ; Tel.: +48-12-664-6180
| |
Collapse
|
38
|
Mesenchymal Stem Cell-Based Therapy as an Alternative to the Treatment of Acute Respiratory Distress Syndrome: Current Evidence and Future Perspectives. Int J Mol Sci 2021; 22:ijms22157850. [PMID: 34360616 PMCID: PMC8346146 DOI: 10.3390/ijms22157850] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) represents a current challenge for medicine due to its incidence, morbidity and mortality and, also, the absence of an optimal treatment. The COVID-19 outbreak only increased the urgent demand for an affordable, safe and effective treatment for this process. Early clinical trials suggest the therapeutic usefulness of mesenchymal stem cells (MSCs) in acute lung injury (ALI) and ARDS. MSC-based therapies show antimicrobial, anti-inflammatory, regenerative, angiogenic, antifibrotic, anti-oxidative stress and anti-apoptotic actions, which can thwart the physiopathological mechanisms engaged in ARDS. In addition, MSC secretome and their derived products, especially exosomes, may reproduce the therapeutic effects of MSC in lung injury. This last strategy of treatment could avoid several safety issues potentially associated with the transplantation of living and proliferative cell populations and may be formulated in different forms. However, the following diverse limitations must be addressed: (i) selection of the optimal MSC, bearing in mind both the heterogeneity among donors and across different histological origins, (ii) massive obtention of these biological products through genetic manipulations of the most appropriate MSC, (iii) bioreactors that allow their growth in 3D, (iv) ideal culture conditions and (v) adequate functional testing of these obtaining biological products before their clinical application.
Collapse
|
39
|
Parsons AM, Darling EM. Temporal responsiveness of adipose-derived stem/stromal cell immune plasticity. Exp Cell Res 2021; 406:112738. [PMID: 34270981 DOI: 10.1016/j.yexcr.2021.112738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/29/2022]
Abstract
We determined the role of time in adipose-derived stem/stromal cell (ASC) response to a model inflammatory environment. ASCs and other mesenchymal stem/stromal cells exhibit immune plasticity. We evaluated the persistence of pro- and anti-inflammatory phenotypes for ASCs exposed to a sustained or pulse inflammatory stimulus. Using qPCR, flow cytometry, and immunocytochemistry, we monitored the temporal expression and up-regulation patterns of a pro-inflammatory gene (caspase 1), a pleiotropic gene/protein (interleukin 6, IL-6), and an anti-inflammatory gene/protein (indoleamine 2, 3-dioxygenase, IDO1) after exposing ASCs to the cytokines tumor necrosis factor-α and interferon-γ. In response to sustained cytokine stimulation, we discovered that time played a role in the balance of pro- and anti-inflammatory ASC phenotypes. IL-6 was present at all time points for both cytokine-stimulated and non-stimulated conditions, whereas IDO1 was heterogeneously up-regulated in stimulated conditions at later time points. After a pulse stimulus, ASC immunoresponse remained consistent for 96-168 h. As a final measure of immune plasticity, we cultured cytokine-stimulated ASCs with blood-derived macrophages to observe macrophage polarization. While the presence of ASCs altered macrophage phenotype, there was no dependency on the length of ASC cytokine exposure time.
Collapse
Affiliation(s)
| | - Eric M Darling
- Department of Pathology and Laboratory Medicine, Brown University, United States; Center for Biomedical Engineering, Brown University, United States; School of Engineering, Brown University, United States; Department of Orthopaedics, Brown University, United States.
| |
Collapse
|
40
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
41
|
Shin EY, Yoon YJ, Lee JE, Shim SH, Park GH, Lee DR. Identification of Putative Markers That Predict the In Vitro Senescence of Mesenchymal Progenitor Cells. Cells 2021; 10:cells10061301. [PMID: 34073789 PMCID: PMC8225148 DOI: 10.3390/cells10061301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal progenitor cells (MPCs) are a promising cell source for regenerative medicine because of their immunomodulatory properties, anti-inflammatory molecule secretion, and replacement of damaged cells. Despite these advantages, heterogeneity in functional potential and limited proliferation capacity of MPCs, as well as the lack of suitable markers for product potency, hamper the development of large-scale manufacturing processes of MPCs. Therefore, there is a sustained need to develop highly proliferative and standardized MPCs in vitro and find suitable functional markers for measuring product potency. In this study, three lines of pluripotent stem cell (PSC)-derived MPCs with high proliferative ability were established and compared with bone-marrow-derived MPCs using proliferation assays and microarrays. A total of six genes were significantly overexpressed (>10-fold) in the highest proliferative MPC line (CHA-hNT5-MPCs) and validated by qRT-PCR. However, only two of the genes (MYOCD and ODZ2) demonstrated a significant correlation with MPC senescence in vitro. Our study provides new gene markers for predicting replicative senescence and the available quantity of MPCs but may also help to guide the development of new standard criteria for manufacturing.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Yeo-Joon Yoon
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, CHA University, Seongnam 13488, Gyunggi-do, Korea;
| | - Sung Han Shim
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Gene Hong Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
- Correspondence:
| |
Collapse
|
42
|
Li L, Peng Y, Yuan Q, Sun J, Zhuang A, Bi X. Cathelicidin LL37 Promotes Osteogenic Differentiation in vitro and Bone Regeneration in vivo. Front Bioeng Biotechnol 2021; 9:638494. [PMID: 34012955 PMCID: PMC8126666 DOI: 10.3389/fbioe.2021.638494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
Different types of biomaterials have been used to repair the defect of bony orbit. However, exposure and infections are still critical risks in clinical application. Biomaterials with characteristics of osteogenesis and antibiosis are needed for bone regeneration. In this study, we aimed to characterize the antimicrobial effects of cathelicidin-LL37 and to assess any impacts on osteogenic activity. Furthermore, we attempted to demonstrate the feasibility of LL37 as a potential strategy in the reconstruction of clinical bone defects. Human adipose-derived mesenchyme stem cells (hADSCs) were cultured with different concentrations of LL37 and the optimum concentration for osteogenesis was selected for further in vitro studies. We then evaluated the antibiotic properties of LL37 at the optimum osteogenic concentration. Finally, we estimated the efficiency of a PSeD/hADSCs/LL37 combined scaffold on reconstructing bone defects in the rat calvarial defect model. The osteogenic ability on hADSCs in vitro was shown to be dependent on the concentration of LL37 and reached a peak at 4 μg/ml. The optimum concentration of LL37 showed good antimicrobial properties against Escherichia coli and Staphylococcus anurans. The combination scaffold of PSeD/hADSCs/LL37 showed superior osteogenic properties compared to the PSeD/hADSCs, PSeD, and control groups scaffolds, indicating a strong bone reconstruction effect in the rat calvarial bone defect model. In Conclusion, LL37 was shown to promote osteogenic differentiation in vitro as well as antibacterial properties. The combination of PSeD/hADSCs/LL37 was advantageous in the rat calvarial defect reconstruction model, showing high potential in clinical bone regeneration.
Collapse
Affiliation(s)
- Lunhao Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yiyu Peng
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qingyue Yuan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jing Sun
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ai Zhuang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xiaoping Bi
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
43
|
Fernández-Francos S, Eiro N, Costa LA, Escudero-Cernuda S, Fernández-Sánchez ML, Vizoso FJ. Mesenchymal Stem Cells as a Cornerstone in a Galaxy of Intercellular Signals: Basis for a New Era of Medicine. Int J Mol Sci 2021; 22:ijms22073576. [PMID: 33808241 PMCID: PMC8036553 DOI: 10.3390/ijms22073576] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Around 40% of the population will suffer at some point in their life a disease involving tissue loss or an inflammatory or autoimmune process that cannot be satisfactorily controlled with current therapies. An alternative for these processes is represented by stem cells and, especially, mesenchymal stem cells (MSC). Numerous preclinical studies have shown MSC to have therapeutic effects in different clinical conditions, probably due to their mesodermal origin. Thereby, MSC appear to play a central role in the control of a galaxy of intercellular signals of anti-inflammatory, regenerative, angiogenic, anti-fibrotic, anti-oxidative stress effects of anti-apoptotic, anti-tumor, or anti-microbial type. This concept forces us to return to the origin of natural physiological processes as a starting point to understand the evolution of MSC therapy in the field of regenerative medicine. These biological effects, demonstrated in countless preclinical studies, justify their first clinical applications, and draw a horizon of new therapeutic strategies. However, several limitations of MSC as cell therapy are recognized, such as safety issues, handling difficulties for therapeutic purposes, and high economic cost. For these reasons, there is an ongoing tendency to consider the use of MSC-derived secretome products as a therapeutic tool, since they reproduce the effects of their parent cells. However, it will be necessary to resolve key aspects, such as the choice of the ideal type of MSC according to their origin for each therapeutic indication and the implementation of new standardized production strategies. Therefore, stem cell science based on an intelligently designed production of MSC and or their derivative products will be able to advance towards an innovative and more personalized medical biotechnology.
Collapse
Affiliation(s)
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216)
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
| | - Sara Escudero-Cernuda
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain; (S.E.-C.); (M.L.F.-S.)
| | - María Luisa Fernández-Sánchez
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain; (S.E.-C.); (M.L.F.-S.)
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, 33290 Gijón, Spain; (S.F.-F.); (L.A.C.)
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-985320050 (ext. 84216)
| |
Collapse
|
44
|
Mesenchymal stromal cells for the treatment of ocular autoimmune diseases. Prog Retin Eye Res 2021; 85:100967. [PMID: 33775824 DOI: 10.1016/j.preteyeres.2021.100967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/22/2022]
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, have emerged as a promising cell-based therapy for a range of autoimmune diseases thanks to several therapeutic advantages. Key among these are: 1) the ability to modulate innate and adaptive immune responses and to promote tissue regeneration, 2) the ease of their isolation from readily accessible tissues and expansion at scale in culture, 3) their low immunogenicity enabling use as an allogeneic "off-the-shelf" product, and 4) MSC therapy's safety and feasibility in humans, as demonstrated in more than one thousand clinical trials. Evidence from preclinical studies and early clinical trials indicate the therapeutic potential of MSCs and their derivatives for efficacy in ocular autoimmune diseases such as autoimmune uveoretinitis and Sjögren's syndrome-related dry eye disease. In this review, we provide an overview of the current understanding of the therapeutic mechanisms of MSCs, and summarize the results from preclinical and clinical studies that have used MSCs or their derivatives for the treatment of ocular autoimmune diseases. We also discuss the challenges to the successful clinical application of MSC therapy, and suggest strategies for overcoming them.
Collapse
|
45
|
Kouroupis D, Correa D. Increased Mesenchymal Stem Cell Functionalization in Three-Dimensional Manufacturing Settings for Enhanced Therapeutic Applications. Front Bioeng Biotechnol 2021; 9:621748. [PMID: 33644016 PMCID: PMC7907607 DOI: 10.3389/fbioe.2021.621748] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/07/2021] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC) exist within their in vivo niches as part of heterogeneous cell populations, exhibiting variable stemness potential and supportive functionalities. Conventional extensive 2D in vitro MSC expansion, aimed at obtaining clinically relevant therapeutic cell numbers, results in detrimental effects on both cellular characteristics (e.g., phenotypic changes and senescence) and functions (e.g., differentiation capacity and immunomodulatory effects). These deleterious effects, added to the inherent inter-donor variability, negatively affect the standardization and reproducibility of MSC therapeutic potential. The resulting manufacturing challenges that drive the qualitative variability of MSC-based products is evident in various clinical trials where MSC therapeutic efficacy is moderate or, in some cases, totally insufficient. To circumvent these limitations, various in vitro/ex vivo techniques have been applied to manufacturing protocols to induce specific features, attributes, and functions in expanding cells. Exposure to inflammatory cues (cell priming) is one of them, however, with untoward effects such as transient expression of HLA-DR preventing allogeneic therapeutic schemes. MSC functionalization can be also achieved by in vitro 3D culturing techniques, in an effort to more closely recapitulate the in vivo MSC niche. The resulting spheroid structures provide spatial cell organization with increased cell–cell interactions, stable, or even enhanced phenotypic profiles, and increased trophic and immunomodulatory functionalities. In that context, MSC 3D spheroids have shown enhanced “medicinal signaling” activities and increased homing and survival capacities upon transplantation in vivo. Importantly, MSC spheroids have been applied in various preclinical animal models including wound healing, bone and osteochondral defects, and cardiovascular diseases showing safety and efficacy in vivo. Therefore, the incorporation of 3D MSC culturing approach into cell-based therapy would significantly impact the field, as more reproducible clinical outcomes may be achieved without requiring ex vivo stimulatory regimes. In the present review, we discuss the MSC functionalization in 3D settings and how this strategy can contribute to an improved MSC-based product for safer and more effective therapeutic applications.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, United States.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, United States.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
46
|
Chieregato K, Bernardi M, Alghisi A, Giordano R, Lazzari L, Perbellini O, Rassu M, Ruggeri M, Astori G. A flow cytometric assay for the quantification of MSC lysis by peripheral blood mononucleated cells. Heliyon 2021; 7:e06036. [PMID: 33553772 PMCID: PMC7856420 DOI: 10.1016/j.heliyon.2021.e06036] [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: 10/23/2020] [Revised: 12/31/2020] [Accepted: 01/14/2021] [Indexed: 11/03/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are attractive candidates for the treatment of acute graft versus host disease (aGvHD) or autoimmune disorders. However, mechanisms of MSC recognition remain unclear and there are evidences that MSC are not totally immunoprivileged. Data suggest that MSC undergo apoptosis after infusion in presence of cytotoxic cells and their death could drive immunosuppression. In GvHD patients, that activity was associated with clinical response. It is mandatory to develop an in vitro potency testing predictor of the "in vivo" response to the therapy. We describe a flow cytometric assay based on differential immunostaining of target and effector cells where BM MSC are enumerated with fluorospheres to determine the loss of target cells after co-culture with PB MNC. 6/13 (46%) of BM MSC lots were lysed by PB MNC and the lysis was proportional to the E/T cell ratio. The method overcomes the problems linked to the use of dyes or radioactive, evidencing the limitations linked to the use of a single vital dye and proposing a precise gating strategy based on absolute cell counts where cells are left untouched. The assay is easy and could be used to predict the response of the patients to the therapy.
Collapse
Affiliation(s)
- Katia Chieregato
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy.,CORIS Veneto - Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy.,CORIS Veneto - Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Alberta Alghisi
- Immunohematology and Transfusion Medicine Service, Vicenza Hospital, Italy
| | - Rosaria Giordano
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Omar Perbellini
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| | - Mario Rassu
- Department of Microbiology, Vicenza Hospital, Italy
| | - Marco Ruggeri
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| |
Collapse
|
47
|
Lee W, Wang LT, Yen ML, Hsu PJ, Lee YW, Liu KJ, Lin KI, Su YW, Sytwu HK, Yen BL. Resident vs nonresident multipotent mesenchymal stromal cell interactions with B lymphocytes result in disparate outcomes. Stem Cells Transl Med 2021; 10:711-724. [PMID: 33506633 PMCID: PMC8046079 DOI: 10.1002/sctm.20-0289] [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: 07/01/2020] [Revised: 12/17/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023] Open
Abstract
Multipotent human mesenchymal stromal cells (MSCs) from multiple organs including the bone marrow (BM) and placenta harbor clinically relevant immunomodulation best demonstrated toward T lymphocytes. Surprisingly, there is limited knowledge on interactions with B lymphocytes, which originate from the BM where there is a resident MSC. With increasing data demonstrating MSC tissue‐specific propensities impacting therapeutic outcome, we therefore investigated the interactions of BM‐MSCs—its resident and “niche” MSC—and placental MSCs (P‐MSCs), another source of MSCs with well‐characterized immunomodulatory properties, on the global functional outcomes of pan‐peripheral B cell populations. We found that P‐MSCs but not BM‐MSCs significantly inhibit proliferation and further differentiation of stimulated human peripheral B populations in vitro. Moreover, although BM‐MSCs preserve multiple IL‐10‐producing regulatory B cell (Breg) subsets, P‐MSCs significantly increase all subsets. To corroborate these in vitro findings in vivo, we used a mouse model of B‐cell activation and found that adoptive transfer of P‐MSCs but not BM‐MSCs significantly decreased activated B220+ B cells. Moreover, adoptive transfer of P‐MSCs but not BM‐MSCs significantly decreased the overall B220+ B‐cell proliferation and further differentiation, similar to the in vitro findings. P‐MSCs also increased two populations of IL‐10‐producing murine Bregs more strongly than BM‐MSCs. Transcriptome analyses demonstrated multifactorial differences between BM‐ and P‐MSCs in the profile of relevant factors involved in B lymphocyte proliferation and differentiation. Our results highlight the divergent outcomes of tissue‐specific MSCs interactions with peripheral B cells, and demonstrate the importance of understanding tissue‐specific differences to achieve more efficacious outcome with MSC therapy.
Collapse
Affiliation(s)
- Wei Lee
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan.,Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, Taiwan
| | - Li-Tzu Wang
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei, Taiwan
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, Taiwan
| | - Yu-Wei Lee
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Tainan, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, NHRI, Zhunan, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases & Vaccinology, NHRI, Zhunan, Taiwan.,Graduate Institute of Microbiology & Immunology, NDMC, Taipei, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, Taiwan
| |
Collapse
|
48
|
Introna M, Golay J. Tolerance to Bone Marrow Transplantation: Do Mesenchymal Stromal Cells Still Have a Future for Acute or Chronic GvHD? Front Immunol 2020; 11:609063. [PMID: 33362797 PMCID: PMC7759493 DOI: 10.3389/fimmu.2020.609063] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal Stromal Cells (MSCs) are fibroblast-like cells of mesodermal origin present in many tissues and which have the potential to differentiate to osteoblasts, adipocytes and chondroblasts. They also have a clear immunosuppressive and tissue regeneration potential. Indeed, the initial classification of MSCs as pluripotent stem cells, has turned into their identification as stromal progenitors. Due to the relatively simple procedures available to expand in vitro large numbers of GMP grade MSCs from a variety of different tissues, many clinical trials have tested their therapeutic potential in vivo. One pathological condition where MSCs have been quite extensively tested is steroid resistant (SR) graft versus host disease (GvHD), a devastating condition that may occur in acute or chronic form following allogeneic hematopoietic stem cell transplantation. The clinical and experimental results obtained have outlined a possible efficacy of MSCs, but unfortunately statistical significance in clinical studies has only rarely been reached and effects have been relatively limited in most cases. Nonetheless, the extremely complex pathogenetic mechanisms at the basis of GvHD, the fact that studies have been conducted often in patients who had been previously treated with multiple lines of therapy, the variable MSC doses and schedules administered in different trials, the lack of validated potency assays and clear biomarkers, the difference in MSC sources and production methods may have been major factors for this lack of clear efficacy in vivo. The heterogeneity of MSCs and their different stromal differentiation potential and biological activity may be better understood through more refined single cell sequencing and proteomic studies, where either an “anti-inflammatory” or a more “immunosuppressive” profile can be identified. We summarize the pathogenic mechanisms of acute and chronic GvHD and the role for MSCs. We suggest that systematic controlled clinical trials still need to be conducted in the most promising clinical settings, using better characterized cells and measuring efficacy with specific biomarkers, before strong conclusions can be drawn about the therapeutic potential of these cells in this context. The same analysis should be applied to other inflammatory, immune or degenerative diseases where MSCs may have a therapeutic potential.
Collapse
Affiliation(s)
- Martino Introna
- Center of Cellular Therapy "G. Lanzani", Division of Haematology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Josée Golay
- Center of Cellular Therapy "G. Lanzani", Division of Haematology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.,Fondazione per la Ricerca Ospedale Maggiore, Bergamo, Italy
| |
Collapse
|
49
|
Gurung S, Ulrich D, Sturm M, Rosamilia A, Werkmeister JA, Gargett CE. Comparing the Effect of TGF-β Receptor Inhibition on Human Perivascular Mesenchymal Stromal Cells Derived from Endometrium, Bone Marrow and Adipose Tissues. J Pers Med 2020; 10:jpm10040261. [PMID: 33271899 PMCID: PMC7712261 DOI: 10.3390/jpm10040261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Rare perivascular mesenchymal stromal cells (MSCs) with therapeutic properties have been identified in many tissues. Their rarity necessitates extensive in vitro expansion, resulting in spontaneous differentiation, cellular senescence and apoptosis, producing therapeutic products with variable quality and decreased potency. We previously demonstrated that A83-01, a transforming growth factor beta (TGF-β) receptor inhibitor, maintained clonogenicity and promoted the potency of culture-expanded premenopausal endometrial MSCs using functional assays and whole-transcriptome sequencing. Here, we compared the effects of A83-01 on MSCs derived from postmenopausal endometrium, menstrual blood, placenta decidua-basalis, bone marrow and adipose tissue. Sushi-domain-containing-2 (SUSD2+) and CD34+CD31−CD45− MSCs were isolated. Expanded MSCs were cultured with or without A83-01 for 7 days and assessed for MSC properties. SUSD2 identified perivascular cells in the placental decidua-basalis, and their maternal origin was validated. A83-01 promoted MSC proliferation from all sources except bone marrow and only increased SUSD2 expression and prevented apoptosis in MSCs from endometrial-derived tissues. A83-01 only improved the cloning efficiency of postmenopausal endometrial MSCs (eMSCs), and expanded adipose tissue MSCs (adMSCs) underwent significant senescence, which was mitigated by A83-01. MSCs derived from bone marrow (bmMSCs) were highly apoptotic, but A83-01 was without effect. A83-01 maintained the function and phenotype in MSCs cultured from endometrial, but not other, tissues. Our results also demonstrated that cellular SUSD2 expression directly correlates with the functional phenotype.
Collapse
Affiliation(s)
- Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
- Correspondence: ; Tel.: +61-03-8572-2813
| | - Daniela Ulrich
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz, 8036 Graz, Austria
| | - Marian Sturm
- Cell & Tissue Therapies WA, Royal Perth Hospital, Perth, WA 6000, Australia;
- Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Anna Rosamilia
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
- Monash Health, Clayton, VIC 3168, Australia
| | - Jerome A. Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (D.U.); (J.A.W.); (C.E.G.)
- Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia;
| |
Collapse
|
50
|
Misiak J, Jean R, Rodriguez S, Deleurme L, Lamy T, Tarte K, Amé-Thomas P. Human Lymphoid Stromal Cells Contribute to Polarization of Follicular T Cells Into IL-4 Secreting Cells. Front Immunol 2020; 11:559866. [PMID: 33133070 PMCID: PMC7562812 DOI: 10.3389/fimmu.2020.559866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Fibroblastic reticular cells (FRCs) are the specialized lymphoid stromal cells initially identified as triggering T-cell recruitment and dynamic motion in secondary lymphoid organs. Interestingly, FRCs also display antigen presentation capacities and support lymphocyte survival. CXCR5+CD4+ follicular T cells are important players of B-cell maturation and antibody response. Our study reported that in vitro-differentiated FRC-like cells enhanced the growth of the whole CXCR5+CD4+ T-cell compartment, while enhancing IL-4 secretion specifically by the PD1dimCXCR5+CD4+ cell subset, in a Notch- and ICAM1/LFA1-dependent manner. In addition, we revealed that in follicular lymphoma (FL) tissues, previously identified as enriched for PD1hiCXCR5hiCD4+ mature follicular helper T cells, PD1dimCXCR5+CD4+ T cells displayed an enrichment for Notch and integrin gene signatures, and a Notch and ICAM-1-dependent overexpression of IL-4 compared to their non-malignant counterparts. These findings suggest that the crosstalk between FRCs and CXCR5+PD1dimCD4+ T cells may contribute to the FL IL-4 rich environment, thus providing new insights in FL lymphomagenesis.
Collapse
Affiliation(s)
- Jan Misiak
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France
| | - Rachel Jean
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France.,CHU de Rennes, Pôle Biologie, Rennes, France
| | - Stéphane Rodriguez
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France
| | - Laurent Deleurme
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France.,Univ Rennes, CNRS, Inserm, BIOSIT (Biologie, Santé, Innovation Technologique de Rennes)-Unité Mixte de Service 3480, Rennes, France
| | - Thierry Lamy
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France.,CHU de Rennes, Service d'Hématologie Clinique, Rennes, France
| | - Karin Tarte
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France.,CHU de Rennes, Pôle Biologie, Rennes, France
| | - Patricia Amé-Thomas
- INSERM U1236, Univ Rennes, Etablissement Français du Sang Bretagne, LabEx IGO, Rennes, France.,CHU de Rennes, Pôle Biologie, Rennes, France
| |
Collapse
|