1
|
Zargarani S, Tavaf MJ, Soltanmohammadi A, Yazdanpanah E, Baharlou R, Yousefi B, Sadighimoghaddam B, Esmaeili SA, Haghmorad D. Adipose-derived mesenchymal stem cells ameliorates experimental autoimmune encephalomyelitis via modulation of Th1/Th17 and expansion of Th2/Treg responses. Cell Biol Int 2024; 48:1124-1137. [PMID: 38741520 DOI: 10.1002/cbin.12171] [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: 08/30/2023] [Revised: 02/03/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
The most common central nervous system (CNS) inflammatory disease is multiple sclerosis (MS), modeled using experimental autoimmune encephalomyelitis (EAE). Mesenchymal stem cells (MSCs) exhibit potent immunomodulatory capabilities, including the suppression of immune cell functions and anti-inflammatory cytokine production. Female C57BL/6 mice (8-10 weeks old) were divided into three groups: 1. Control, 2. Allogeneic MSCs (ALO) treatment, and 3. Syngeneic MSCs (SYN) treatment. To induce EAE, myelin oligodendrocyte glycoprotein was injected subcutaneously with complete Freund's adjuvant, followed by intraperitoneal pertussis toxin. On Days 6 and 12 postimmunization, the treatment groups received intraperitoneal injections of 2 × 106 MSCs. Daily clinical and weight assessments were performed, and on Day 25, the mice were euthanized. At the end of the period, brain histological analysis was conducted to quantify lymphocyte infiltration. T-cell characteristics were determined using enzyme-linked immunosorbent assay and Real-time polymerase chain reaction (RT-PCR). The assessment of transcription factor expression levels in the CNS was also performed using RT-PCR. Compared to the control group, both the allogeneic (ALO) and syngeneic (SYN) groups demonstrated significantly reduced disease progression. The maximum clinical scores for the control, ALO, and SYN groups were 4.4 ± 0.1, 2.4 ± 0.2, and 2.1 ± 0.2, respectively (ALO and SYN vs. Control: p < .001). In comparison to the control group, histological studies demonstrated that the allogeneic and syngeneic groups had less lymphocytic infiltration (ALO: 1.4 ± 0.1, SYN: 1.2 ± 0.2, and control: 2.8 ± 0.15; p < .001) and demyelination (ALO: 1.2 ± 0.15, SYN: 1.1 ± 0.1 and control: 2.9 ± 0.1, p < .001). ALO and SYN groups had lower expression of Th1 and Th17 cytokines and transcription factors (IFN-γ: 0.067, 0.051; STAT4: 0.189, 0.162; T-bet: 0.175, 0.163; IL-17: 0.074, 0.061; STAT3: 0.271, 0.253; ROR-γt: 0.163, 0.149, respectively) compared to the control group on Day 25 following EAE induction. Additionally, ALO and SYN groups compared to the control group, expressed more Th2 and Treg cytokines and transcription factors (IL-4: 4.25, 4.63; STAT6: 2.78, 2.96; GATA3: 2.91, 3.08; IL-27: 2.32, 2.46, IL-33: 2.71, 2.85; TGF-β: 4.8, 5.05; IL-10: 4.71, 4.93; CTLA-4: 7.72, 7.95; PD1: 4.12,4.35; Foxp3: 3.82,4.08, respectively). This research demonstrated that MSCs possess the potential to be a therapeutic option for MS and related CNS inflammatory disorders. Their immunomodulatory properties, coupled with the observed reductions in disease severity, lymphocytic infiltration, and demyelination, indicate that MSCs could play a crucial role in altering the course of MS by mitigating inflammatory immune responses and promoting regulatory immune processes. These findings open up new possibilities for the development of MSC-based therapies for MS, and further investigation and clinical trials may be warranted to explore their efficacy and safety in human patients.
Collapse
Affiliation(s)
- Simin Zargarani
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam J Tavaf
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Azita Soltanmohammadi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Bizhan Sadighimoghaddam
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
2
|
Zhang Q, Zeng Z, Wei N, Su Y, Wang J, Ni Q, Wang Y, Yang J, Liu X, Xu H, Wang G, Shan Y, Zhou F. Mesenteric lymph nodes: a critical site for the up-regulatory effect of hUC-MSCs on Treg cells by producing TGF-β1 in colitis treatment. Stem Cell Res Ther 2024; 15:190. [PMID: 38956621 PMCID: PMC11218300 DOI: 10.1186/s13287-024-03809-x] [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: 02/27/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) demonstrate a wide range of therapeutic capabilities in the treatment of inflammatory bowel disease (IBD). The intraperitoneal injection of MSCs has exhibited superior therapeutic efficacy on IBD than intravenous injection. Nevertheless, the precise in vivo distribution of MSCs and their biological consequences following intraperitoneal injection remain inadequately understood. Additional studies are required to explore the correlation between MSCs distribution and their biological effects. METHODS First, the distribution of human umbilical cord MSCs (hUC-MSCs) and the numbers of Treg and Th17 cells in mesenteric lymph nodes (MLNs) were analyzed after intraperitoneal injection of hUC-MSCs. Subsequently, the investigation focused on the levels of transforming growth factor beta1 (TGF-β1), a key cytokine to the biology of both Treg and Th17 cells, in tissues of mice with colitis, particularly in MLNs. The study also delved into the impact of hUC-MSCs therapy on Treg cell counts in MLNs, as well as the consequence of TGFB1 knockdown hUC-MSCs on the differentiation of Treg cells and the treatment of IBD. RESULTS The therapeutic effectiveness of intraperitoneally administered hUC-MSCs in the treatment of colitis was found to be significant, which was closely related to their quick migration to MLNs and secretion of TGF-β1. The abundance of hUC-MSCs in MLNs of colitis mice is much higher than that in other organs even the inflamed sites of colon. Intraperitoneal injection of hUC-MSCs led to a significant increase in the number of Treg cells and a decrease in Th17 cells especially in MLNs. Furthermore, the concentration of TGF-β1, the key cytokine for Treg differentiation, were also found to be significantly elevated in MLNs after hUC-MSCs treatment. Knockdown of TGFB1 in hUC-MSCs resulted in a noticeable reduction of Treg cells in MLNs and the eventually failure of hUC-MSCs therapy in colitis. CONCLUSIONS MLNs may be a critical site for the regulatory effect of hUC-MSCs on Treg/Th17 cells and the therapeutic effect on colitis. TGF-β1 derived from hUC-MSCs promotes local Treg differentiation in MLNs. This study will provide new ideas for the development of MSC-based therapeutic strategies in IBD patients.
Collapse
Affiliation(s)
- Qixiang Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhu Zeng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Yueyan Su
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Qi Ni
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yukai Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jingwen Yang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaoyan Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huanke Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- , No. 639 Longmian Avenue, Nanjing, Jiangsu, China.
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Tongjiaxiang #24, Nanjing, Jiangsu, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- , No. 639 Longmian Avenue, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Kobayashi K, Maeda T, Ayodeji M, Tu SC, Chen A, Rajtboriraks M, Hsu CH, Tu TW, Wang PC, Hanley PJ, Jonas RA, Ishibashi N. Dose Effect of Mesenchymal Stromal Cell Delivery Through Cardiopulmonary Bypass. Ann Thorac Surg 2023; 116:1337-1345. [PMID: 35952858 PMCID: PMC10009803 DOI: 10.1016/j.athoracsur.2022.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/22/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neurologic impairments are a significant concern for survivors after pediatric cardiac surgery with cardiopulmonary bypass (CPB). We have previously shown that mesenchymal stromal cell (MSC) delivery through CPB has the potential to mitigate the effects of CPB on neural stem/progenitor cells. This study assessed the dose effects of MSCs. METHODS Piglets (n = 20) were randomly assigned to 1 of 4 groups: control, CPB, or CPB followed by MSC administration with low and high doses (10 × 106 and 100 × 106 cells per kilogram). We assessed acute dose effect on cell distribution, multiorgan functions, systemic inflammation, microglia activation, and neural stem/progenitor cell activities. RESULTS By magnetic resonance imaging, approximately 10 times more MSCs were detected within the entire brain after high-dose delivery than after low-dose delivery. No adverse events affecting hemodynamics, various biomarkers, and neuroimaging were detected after high-dose MSC delivery. High-dose MSCs significantly increased circulating levels of interleukin 4 after CPB. Both MSC groups normalized microglia activation after CPB, demonstrating MSC-induced reduction in cerebral inflammation. There was a significant increase in neuroblasts in the subventricular zone in both treatment groups. The thickness of the most active neurogenic area within the subventricular zone was significantly increased after high-dose treatment compared with CPB and low-dose MSCs, suggesting dose-dependent effects on the neurogenic niche. CONCLUSIONS MSC delivery through CPB is feasible up to 100 × 106 cells per kilogram. MSC treatment during cardiac surgery has the potential to reduce systemic and cerebral inflammation and to modulate responses of an active neurogenic niche to CPB. Further investigation is necessary to assess the long-term effects and to develop a more complete dose-response curve.
Collapse
Affiliation(s)
- Kei Kobayashi
- Department of Cardiac Surgery, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC
| | - Takuya Maeda
- Department of Cardiac Surgery, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC
| | - Mobolanle Ayodeji
- Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC
| | - Shao Ching Tu
- Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Alice Chen
- Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC; George Washington University, School of Medicine and Health Sciences, George Washington University, Washington, DC
| | - May Rajtboriraks
- Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC; Department of Biomedical Engineering, The Catholic University of America, Washington, DC
| | - Chao-Hsiung Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC
| | - Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Paul C Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC; Department of Electrical Engineering, Fu Jen Catholic University, Taipei, Taiwan
| | - Patrick J Hanley
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC; Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC
| | - Richard A Jonas
- Department of Cardiac Surgery, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Nobuyuki Ishibashi
- Department of Cardiac Surgery, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC; Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC.
| |
Collapse
|
4
|
Asserson DB. Allogeneic Mesenchymal Stem Cells After In Vivo Transplantation: A Review. Cell Reprogram 2023; 25:264-276. [PMID: 37971885 DOI: 10.1089/cell.2023.0084] [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] [Indexed: 11/19/2023] Open
Abstract
Autologous mesenchymal stem cells (MSCs) are ideal for tissue regeneration because of their ability to circumvent host rejection, but their procurement and processing present logistical and time-sensitive challenges. Allogeneic MSCs provide an alternative cell-based therapy capable of positively affecting all human organ systems, and can be readily available. Extensive research has been conducted in the treatment of autoimmune, degenerative, and inflammatory diseases with such stem cells, and has demonstrated predominantly safe outcomes with minimal complications. Nevertheless, continued clinical trials are necessary to ascertain optimal harvest and transplant techniques.
Collapse
Affiliation(s)
- Derek B Asserson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Pastor JC, Pastor-Idoate S, López-Paniagua M, Para M, Blazquez F, Murgui E, García V, Coco-Martín RM. Intravitreal allogeneic mesenchymal stem cells: a non-randomized phase II clinical trial for acute non-arteritic optic neuropathy. Stem Cell Res Ther 2023; 14:261. [PMID: 37735668 PMCID: PMC10512539 DOI: 10.1186/s13287-023-03500-7] [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: 02/28/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND An effective treatment for acute non-arteritic ischemic optic neuropathy (NA-AION) has not been known or proven yet. Previous studies have suggested a neuroprotective effect of allogeneic bone marrow-derived mesenchymal stem cells. This study aims to report the results of a clinical trial on patients with acute non-arteritic optic neuropathy (NA-AION) treated with an intravitreal injection of allogeneic bone marrow-derived mesenchymal stem cells (BM-MSCs) (MSV®). METHODS We conducted a prospective, non-randomized, clinical phase-II study (Eudra CT number 2016-003029-40; ClinicalTrials.gov Registry NCT03173638) that included 5 patients with acute unilateral NA-AION diagnosed within 2 weeks after symptom onset and who received an intravitreal injection of allogeneic BM-MSCs (0.05 ml; cell concentration: 1.5 × 106cells/mL). The patients underwent regular ophthalmological examinations and were followed for one year. RESULTS In this trial, allogeneic BM-MSCs appeared to be safe as no patients developed signs of acute nor chronic intraocular inflammation or a significant change in intraocular pressure, although an epiretinal membrane was developed in one patient. A retrolental aggregate formed shortly after the injection spontaneously disappeared within a few weeks in another phakic patient, leaving a subcapsular cataract. Visual improvement was noted in 4 patients, and amplitudes of P100 on the visually evoked potentials recordings increased in three patients. The retinal nerve fiber layer and macular ganglion cell layer thicknesses significantly decreased during the follow-up. CONCLUSIONS Besides the development of an epiretinal membrane in one patient, the intravitreal application of allogeneic BM-MSCs appeared to be intraocularly well tolerated. Consequently, not only NA-AION but also BM-MSCs deserve more clinical investigational resources and a larger randomized multicenter trial that would provide stronger evidence both about safety and the potential therapeutic efficacy of intravitreally injected allogeneic BM-MSCs in acute NA-AION. TRIAL REGISTRATION Safety Assessment of Intravitreal Mesenchymal Stem Cells for Acute Non-Arteritic Anterior Ischemic Optic Neuropathy (NEUROSTEM). NCT03173638. Registered June 02, 2017 https://clinicaltrials.gov/ct2/show/NCT03173638 .
Collapse
Affiliation(s)
- Jose C Pastor
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain
- Hospital Clínico Universitario, Valladolid, Spain
| | - Salvador Pastor-Idoate
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain.
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain.
- Hospital Clínico Universitario, Valladolid, Spain.
| | - Marina López-Paniagua
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Para
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
| | - Francisco Blazquez
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Murgui
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
| | - Verónica García
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Citospin S.L., Valladolid, Spain
| | - Rosa M Coco-Martín
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Pº de Belén nº 17, 47011, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Riekert M, Almanzar G, Schmalzing M, Schütze N, Jakob F, Prelog M. Mesenchymal stem cells modulate IL-17 and IL-9 production induced by Th17-inducing cytokine conditions in autoimmune arthritis: an explorative analysis. Adv Rheumatol 2023; 63:37. [PMID: 37525265 DOI: 10.1186/s42358-023-00317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/19/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND The importance of proinflammatory T-cells and their cytokine production in patients with autoimmune arthritis has been widely described. Due to their immunomodulatory properties, mesenchymal stem cells (MSCs) have come into focus as a potential therapeutic concept. The aim of this study was to investigate the influence of MSCs on the phenotype, cytokine profile, and functionality of naive and non-naive CD4+ T-cells from healthy donors (HD) and patients with autoimmune arthritis under Th17-cytokine polarizing conditions in an explorative way using a transwell system prohibiting any cell-cell-contact. METHODS Magnetically isolated naive and non-naive CD4+ T-cells were stimulated under Th17-polarizing proinflammatory cytokine conditions in presence and absence of bone marrow derived mesenchymal stromal cells (MSCs). After an incubation period of 6 days, the proportions of the T-cell subpopulations TEMRA (CD45RA+CD27-), memory (CD45RA-CD27+), effector (CD45RA-CD27-) and naive cells (CD45RA+CD27+) were determined. Quantitative immunofluorescence intensity was used as a measure for IL-9, IL-17 and IFN-γ production in each subpopulation. RESULTS In isolated naive CD4+ T-cells from HD and patients, MSCs suppressed the differentiation of naive towards an effector phenotype while memory and naive cells showed higher percentages in culture with MSCs. In patients, MSCs significantly decreased the proportion of IL-9 and IL-17 producing effector T-cells. MSCs also reduced IFN-γ production in the naive and memory phenotype from HD. CONCLUSION The results of the study indicate significant immunomodulatory properties of MSCs, as under Th17-polarizing conditions MSCs are still able to control T-cell differentiation and proinflammatory cytokine production in both HD and patients with autoimmune arthritis.
Collapse
Affiliation(s)
- Maximilian Riekert
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany.
- Department of Oral and Craniomaxillofacial and Plastic Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50924, Cologne, Germany.
| | - Giovanni Almanzar
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marc Schmalzing
- Department of Internal Medicine II, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Norbert Schütze
- Orthopedic Clinic, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Clinic, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Prelog
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
7
|
Schlegel P, Yan K, Upadhyaya S, Buyens W, Wong K, Chen A, Faull KF, Al-Hiyari Y, Long J. Tissue-engineered vocal fold replacement in swine: Methods for functional and structural analysis. PLoS One 2023; 18:e0284135. [PMID: 37083641 PMCID: PMC10120936 DOI: 10.1371/journal.pone.0284135] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
We have developed a cell-based outer vocal fold replacement (COVR) as a potential therapy to improve voice quality after vocal fold (VF) injury, radiation, or tumor resection. The COVR consists of multipotent human adipose-derived stem cells (hASC) embedded within a three-dimensional fibrin scaffold that resembles vocal fold epithelium and lamina propria layers. Previous work has shown improved wound healing in rabbit studies. In this pilot study in pigs, we sought to develop methods for large animal implantation and phonatory assessment. Feasibility, safety, and structural and functional outcomes of the COVR implant are described. Of eight pigs studied, six animals underwent COVR implantation with harvest between 2 weeks and 6 months. Recovery of laryngeal tissue structure was assessed by vibratory and histologic analyses. Recovery of voice function was assessed by investigating acoustic parameters that were derived specifically for pigs. Results showed improved lamina propria qualities relative to an injured control animal at 6 months. Acoustic parameters reflected voice worsening immediately after surgery as expected; acoustics displayed clear voice recovery in the animal followed for 6 months after COVR. These methods form the basis for a larger-scale long-term pre-clinical safety and efficacy study.
Collapse
Affiliation(s)
- Patrick Schlegel
- Department of Head and Neck Surgery, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Kenneth Yan
- Department of Otolaryngology/Head and Neck Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Sreenivasa Upadhyaya
- Department of Computer Science, Katholieke Universiteit Leuven, Leuven, Belgium
- SoundTalks N.V, Leuven, Belgium
| | | | - Kirsten Wong
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Anthony Chen
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Kym F. Faull
- Department of Psychiatry & Biobehavioral Sciences, Pasarow Mass Spectrometry Laboratory, Jane & Terry Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Yazeed Al-Hiyari
- Department of Head and Neck Surgery, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Long
- Department of Head and Neck Surgery, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, United States of America
- Surgery and Perioperative Careline, Greater Los Angeles VA Healthcare System, Los Angeles, California, United States of America
| |
Collapse
|
8
|
Wang G, Wang W. Advanced Cell Therapies for Glioblastoma. Front Immunol 2022; 13:904133. [PMID: 36052072 PMCID: PMC9425637 DOI: 10.3389/fimmu.2022.904133] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
The sheer ubiquity of Gioblastoma (GBM) cases would lead you to believe that there should have been many opportunities for the discovery of treatments to successfully render it into remission. Unfortunately, its persistent commonality is due in large part to the fact that it is the most treatment-resistant tumors in adults. That completely changes the treatment plan of attack. Long established and accepted treatment therapies such as surgical resection, radiation, and aggressive chemotherapy, (and any combination thereof) have only confirmed that the disease lives up to its treatment-resistant reputation. To add to the seemingly insurmountable task of finding a cure, GBM has also proven to be a very stubborn and formidable opponent to newer immunotherapies. Across the board, regardless of the therapy combination, the five-year survival rate of GBM patients is still very poor at a heartbreaking 5.6%. Obviously, the present situation cannot be tolerated or deemed acceptable. The grave situation calls for researchers to be more innovative and find more efficient strategies to discover new and successful strategies to treat GBM. Inspired by researchers worldwide attempting to control GBM, we provide in this review a comprehensive overview of the many diverse cell therapies currently being used to treat GBM. An overview of the treatments include: CAR T cells, CAR NK cells, gamma-delta T cells, NKT cells, dendritic cells, macrophages, as well stem cell-based strategies. To give you the complete picture, we will discuss the efficacy, safety, and developmental stages, the mechanisms of action and the challenges of each of these therapies and detail their potential to be the next-generation immunotherapeutic to eliminate this dreadful disease.
Collapse
Affiliation(s)
- Guangwen Wang
- BlueRock Therapeutics, Department of Process Development, Cambridge, MA, United States
- *Correspondence: Wenshi Wang, ; Guangwen Wang,
| | - Wenshi Wang
- Metagenomi Inc., Department of Cell Therapy, Emeryville, CA, United States
- *Correspondence: Wenshi Wang, ; Guangwen Wang,
| |
Collapse
|
9
|
Song Y, Li R, Ye M, Pan C, Zheng L, Wang ZW, Zhu X. Differences in chemotaxis of human mesenchymal stem cells and cervical cancer cells. Apoptosis 2022; 27:840-851. [PMID: 35849265 DOI: 10.1007/s10495-022-01749-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
Abstract
In the last decade, there has been a rapid expansion in tumor targeted therapy using mesenchymal stem cells (MSCs) based on their unique tropism towards cancer cells. Despite similarities in morphology, immunophenotype, and differential potent in vitro, MSCs originated from different tissues do not necessarily have equivalent biological behaviors. It is important to screen the most chemotactic MSCs to cancer cells. In this study, different MSCs were isolated from various human tissues including adipose, umbilical cord, amniotic membrane, and chorion. The chemotaxis of human MSCs to cervical cancer cells was measured by CCK-8, ELISA and Transwell invasion assays. Western blotting was performed to explore the underlying mechanisms. MSCs derived from distinct sources can be differently recruited to cervical cancer cells, among which chorion-derived MSC (CD-MSC) possessed the strongest tropic capacity. CXCL12 was found to be highly secreted by cervical cancer cells, in parallel with the expression of CXCR4 in all MSCs. CD-MSC displayed the highest level of CXCR4. These results indicated that CXCL12/CXCR4 pathway contributed to the different chemotaxis to cervical cancer cells of each MSCs. This study proposed that CD-MSC with the highest CXCR4 expression is a promising therapeutic vehicle for targeted therapy in cervical cancer.
Collapse
Affiliation(s)
- Yizuo Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Miaomiao Ye
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Chunyu Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Lihong Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
10
|
Keshavarz Shahbaz S, Mansourabadi AH, Jafari D. Genetically engineered mesenchymal stromal cells as a new trend for treatment of severe acute graft-versus-host disease. Clin Exp Immunol 2022; 208:12-24. [PMID: 35274673 PMCID: PMC9113247 DOI: 10.1093/cei/uxac016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/25/2021] [Accepted: 02/07/2022] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a population of non-hematopoietic and self-renewing cells characterized by the potential to differentiate into different cell subtypes. MSCs have interesting features which have attracted a lot of attention in various clinical investigations. Some basic features of MSCs are including the weak immunogenicity (absence of MHC-II and costimulatory ligands accompanied by the low expression of MHC-I) and the potential of plasticity and multi-organ homing via expressing related surface molecules. MSCs by immunomodulatory effects could also ameliorate several immune-pathological conditions like graft-versus-host diseases (GVHD). The efficacy and potency of MSCs are the main objections of MSCs therapeutic applications. It suggested that improving the MSC immunosuppressive characteristic via genetic engineering to produce therapeutic molecules consider as one of the best options for this purpose. In this review, we explain the functions, immunologic properties, and clinical applications of MSCs to discuss the beneficial application of genetically modified MSCs in GVHD.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Science, Qazvin, Iran
| | - Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Davood Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Zanjan, Iran
| |
Collapse
|
11
|
Tevlin R, desJardins-Park H, Huber J, DiIorio S, Longaker M, Wan D. Musculoskeletal tissue engineering: Adipose derived stromal cell implementation for the treatment of osteoarthritis. Biomaterials 2022; 286:121544. [DOI: 10.1016/j.biomaterials.2022.121544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 06/23/2021] [Accepted: 09/13/2021] [Indexed: 11/02/2022]
|
12
|
Mallis P, Chatzistamatiou T, Dimou Z, Sarri EF, Georgiou E, Salagianni M, Triantafyllia V, Andreakos E, Stavropoulos-Giokas C, Michalopoulos E. Mesenchymal stromal cell delivery as a potential therapeutic strategy against COVID-19: Promising evidence from in vitro results. World J Biol Chem 2022; 13:47-65. [PMID: 35432769 PMCID: PMC8966500 DOI: 10.4331/wjbc.v13.i2.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/28/2021] [Accepted: 03/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic, which was initiated in December 2019. COVID-19 is characterized by a low mortality rate (< 6%); however, this percentage is higher in elderly people and patients with underlying disorders. COVID-19 is characterized by mild to severe outcomes. Currently, several therapeutic strategies are evaluated, such as the use of anti-viral drugs, prophylactic treatment, monoclonal antibodies, and vaccination. Advanced cellular therapies are also investigated, thus representing an additional therapeutic tool for clinicians. Mesenchymal stromal cells (MSCs), which are known for their immunoregulatory properties, may halt the induced cytokine release syndrome mediated by SARS-CoV-2, and can be considered as a potential stem cell therapy.
AIM To evaluate the immunoregulatory properties of MSCs, upon stimulation with COVID-19 patient serum.
METHODS MSCs derived from the human Wharton’s Jelly (WJ) tissue and bone marrow (BM) were isolated, cryopreserved, expanded, and defined according to the criteria outlined by the International Society for Cellular Therapies. Then, WJ and BM-MSCs were stimulated with a culture medium containing 15% COVID-19 patient serum, 1% penicillin-streptomycin, and 1% L-glutamine for 48 h. The quantification of interleukin (IL)-1 receptor a (Ra), IL-6, IL-10, IL-13, transforming growth factor (TGF)-β1, vascular endothelial growth factor (VEGF)-a, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and indoleamine-2,3-dioxygenase (IDO) was performed using commercial ELISA kits. The expression of HLA-G1, G5, and G7 was evaluated in unstimulated and stimulated WJ and BM-MSCs. Finally, the interactions between MSCs and patients’ macrophages were established using co-culture experiments.
RESULTS Thawed WJ and BM-MSCs exhibited a spindle-shaped morphology, successfully differentiated to “osteocytes”, “adipocytes”, and “chondrocytes”, and in flow cytometric analysis were characterized by positivity for CD73, CD90, and CD105 (> 95%) and negativity for CD34, CD45, and HLA-DR (< 2%). Moreover, stimulated WJ and BM-MSCs were characterized by increased cytoplasmic granulation, in comparison to unstimulated cells. The HLA-G isoforms (G1, G5, and G7) were successfully expressed by the unstimulated and stimulated WJ-MSCs. On the other hand, only weak expression of HLA-G1 was identified in BM-MSCs. Stimulated MSCs secreted high levels of IL-1Ra, IL-6, IL-10, IL-13, TGF-β1, FGF, VEGF, PDGF, and IDO in comparison to unstimulated cells (P < 0.05) after 12 and 24 h. Finally, macrophages derived from COVID-19 patients successfully adapted the M2 phenotype after co-culturing with stimulated WJ and BM-MSCs.
CONCLUSION WJ and BM-MSCs successfully produced high levels of immunoregulatory agents, which may efficiently modulate the over-activated immune responses of critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | | | - Zetta Dimou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Eirini-Faidra Sarri
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Eleni Georgiou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Maria Salagianni
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | | | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| |
Collapse
|
13
|
Laranjeira P, Pedrosa M, Duarte C, Pedreiro S, Antunes B, Ribeiro T, dos Santos F, Martinho A, Fardilha M, Domingues MR, Abecasis M, Pereira da Silva JA, Paiva A. Human Bone Marrow Mesenchymal Stromal/Stem Cells Regulate the Proinflammatory Response of Monocytes and Myeloid Dendritic Cells from Patients with Rheumatoid Arthritis. Pharmaceutics 2022; 14:pharmaceutics14020404. [PMID: 35214136 PMCID: PMC8880255 DOI: 10.3390/pharmaceutics14020404] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a disabling autoimmune disease whose treatment is ineffective for one-third of patients. Thus, the immunomodulatory potential of mesenchymal stromal/stem cells (MSCs) makes MSC-based therapy a promising approach to RA. This study aimed to explore the immunomodulatory action of human bone marrow (BM)-MSCs on myeloid dendritic cells (mDCs) and monocytes, especially on cytokines/chemokines involved in RA physiopathology. For that, LPS plus IFNγ-stimulated peripheral blood mononuclear cells from RA patients (n = 12) and healthy individuals (n = 6) were co-cultured with allogeneic BM-MSCs. TNF-α, CD83, CCR7 and MIP-1β protein levels were assessed in mDCs, classical, intermediate, and non-classical monocytes. mRNA expression of other cytokines/chemokines was also evaluated. BM-MSCs effectively reduced TNF-α, CD83, CCR7 and MIP-1β protein levels in mDCs and all monocyte subsets, in RA patients. The inhibition of TNF-α production was mainly achieved by the reduction of the percentage of cellsproducing this cytokine. BM-MSCs exhibited a remarkable suppressive action over antigen-presenting cells from RA patients, potentially affecting their ability to stimulate the immune adaptive response at different levels, by hampering their migration to the lymph node and the production of proinflammatory cytokines and chemokines. Accordingly, MSC-based therapies can be a valuable approach for RA treatment, especially for non-responder patients.
Collapse
Affiliation(s)
- Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Av. Bissaya Barreto, Bloco de Celas, 3000-075 Coimbra, Portugal;
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Polo 1, 1.° Piso, FMUC, Rua Larga, 3004-504 Coimbra, Portugal
| | - Mónia Pedrosa
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
- Enzifarma—Diagnostica e Farmacêutica, S.A., Estrada da Luz, n.° 90, 2° F, 1600-160 Lisbon, Portugal
| | - Cátia Duarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Praceta Professor Mota Pinto, 3000-075 Coimbra, Portugal
| | - Susana Pedreiro
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
| | - Brígida Antunes
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
| | - Tânia Ribeiro
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
| | - Francisco dos Santos
- Cell2B Advanced Therapeutics, SA, Biocant Park, Núcleo 04, Lote 4A, 3060-197 Cantanhede, Portugal; ; (B.A.); (T.R.); (F.d.S.)
- Stemlab SA, Biocant Park, Núcleo 04, Lote 2, 3060-197 Cantanhede, Portugal
| | - António Martinho
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
| | - Margarida Fardilha
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
- Laboratory of Signal Transduction, Institute of Biomedicine—iBiMED, Department of Medical Sciences, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - M. Rosário Domingues
- Mass Spectrometry Centre, LAQV REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal;
- Centre for Environmental and Marine Studies (CESAM), Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Manuel Abecasis
- Serviço de Transplantação de Progenitores Hematopoiéticos (UTM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Professor Lima Basto, 1099-023 Lisbon, Portugal;
- Instituto Português do Sangue e da Transplantação—CEDACE, Alameda das Linhas de Torres, 117, 1769-001 Lisbon, Portugal
| | - José António Pereira da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Praceta Professor Mota Pinto, 3000-075 Coimbra, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Av. Bissaya Barreto, Bloco de Celas, 3000-075 Coimbra, Portugal;
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal, Quinta da Vinha Moura, São Martinho do Bispo, 3041-861 Coimbra, Portugal; (M.P.); (S.P.); (A.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (C.D.); (J.A.P.d.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-488-700
| |
Collapse
|
14
|
Chu CF, Mao SH, Shyu VBH, Chen CH, Chen CT. Allogeneic Bone-Marrow Mesenchymal Stem Cell with Moldable Cryogel for Craniofacial Bone Regeneration. J Pers Med 2021; 11:jpm11121326. [PMID: 34945798 PMCID: PMC8704672 DOI: 10.3390/jpm11121326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/20/2022] Open
Abstract
Allogeneic bone-marrow mesenchymal stem cells (BMSCs) can promote bone regeneration and substitute for autologous BMSCs if autologous sources are unavailable, but the efficacy of bone regeneration by allogeneic BMSCs is still inconsistent. A Lewis rat cranium defect model was used to investigate the efficacy of bone regeneration between autologous and allogeneic BMSCs in gelatin-nanohydroxyapatite cryogel scaffolds. BMSCs from Wistar rats served as the allogeneic cell lineage. The full-thickness cranium defects were treated by either blank control, cryogel only, allogeneic BMSC-seeded cryogel, or autologous BMSC-seeded cryogel (n = 5). Bone regeneration was monitored by micro-computed tomography and examined histologically at week 12. In addition, we assessed the immune responses in vitro by mixed lymphocyte reaction (MLR) assay and CD4+ immunochemistry staining ex vivo. The MLR showed that allogeneic BSMCs elicited a weak immune response on day 14 that progressively attenuated by day 28. In vivo, the bone regeneration in allogeneic BMSCs was inferior at week 4, but progressively matched the autologous BMSCs by week 12. Our results suggest that allogeneic BMSCs can serve as an alternative source for bone regeneration.
Collapse
Affiliation(s)
- Cheng-Feng Chu
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-F.C.); (V.B.-H.S.); (C.-H.C.)
| | - Shih-Hsuan Mao
- Department of Plastic and Reconstructive Surgery, College of Medicine, Chang Gung University, Linkou Chang Gung Memorial Hospital, Craniofacial Research Center, Taoyuan 333, Taiwan;
| | - Victor Bong-Hang Shyu
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-F.C.); (V.B.-H.S.); (C.-H.C.)
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-F.C.); (V.B.-H.S.); (C.-H.C.)
| | - Chien-Tzung Chen
- Department of Plastic and Reconstructive Surgery, College of Medicine, Chang Gung University, Linkou Chang Gung Memorial Hospital, Craniofacial Research Center, Taoyuan 333, Taiwan;
- Correspondence: ; Fax: +886-3328-7200
| |
Collapse
|
15
|
Abu-Shahba N, Mahmoud M, El-Erian AM, Husseiny MI, Nour-Eldeen G, Helwa I, Amr K, ElHefnawi M, Othman AI, Ibrahim SA, Azmy O. Impact of type 2 diabetes mellitus on the immunoregulatory characteristics of adipose tissue-derived mesenchymal stem cells. Int J Biochem Cell Biol 2021; 140:106072. [PMID: 34455058 DOI: 10.1016/j.biocel.2021.106072] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/02/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder associated with several complications. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) represent an emerging type of MSCs with high plasticity and immunoregulatory capabilities and are useful for treating inflammation-related disorders such as T2DM. However, the pathogenic microenvironment of T2DM may affect their therapeutic potential. We aimed to examine the impact of the diabetic milieu on the immunomodulatory/anti-inflammatory potential of AT-MSCs. METHODS We assessed the proliferation potential, cell surface expression of MSC-characteristic markers and immunomodulatory markers, along with the gene expression and protein secretion of pro-inflammatory and anti-inflammatory cytokines and adipokines in AT-MSCs derived from T2DM patients (dAT-MSCs) vs. those derived from non-diabetic volunteers (ndAT-MSCs). Furthermore, we evaluated the IFN-γ priming effect on both groups. RESULTS Our data revealed comparable proliferative activities in both groups. Flow cytometric analysis results showed a lower expression of CD200 and CD276 on dAT-MSCs vs. ndAT-MSCs. qPCR demonstrated upregulation of IL-1β associated with a downregulation of IL-1RN in dAT-MSCs vs. ndAT-MSCs. IFN-γ priming induced an elevation in CD274 expression associated with IDO1 and ILRN overexpression and IL-1β downregulation in both groups. ELISA analysis uncovered elevated levels of secreted IL-1β, TNF, and visfatin/NAMPT in dAT-MSCs, whereas IL-1RA and IDO levels were reduced. ELISA results were also evident in the secretome of dAT-MSCs upon IFN-γ priming. CONCLUSIONS This study suggests that the T2DM milieu alters the immunomodulatory characteristics of AT-MSCs with a shift towards a proinflammatory phenotype which may restrain their autologous therapeutic use. Furthermore, our findings indicate that IFN-γ priming could be a useful strategy for enhancing dAT-MSC anti-inflammatory potential.
Collapse
Affiliation(s)
- Nourhan Abu-Shahba
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt; Department of Medical Molecular Genetics, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt.
| | - Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt; Department of Medical Molecular Genetics, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Alaa Mohammed El-Erian
- Department of Endocrine Surgery, National Institute of Diabetes and Endocrinology, Cairo, Egypt
| | - Mohamed Ibrahim Husseiny
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs DMRI, Beckman Research Institute, City of Hope, National Medical Center, Durate, CA, USA; Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ghada Nour-Eldeen
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt; Department of Molecular Genetics and Enzymology, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Iman Helwa
- Department of Immunogenetics, Human Genetics and Genome Research Division, National Resrearch Centre, Egypt
| | - Khalda Amr
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Mahmoud ElHefnawi
- Biomedical Informatics and Chemoinformatics Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt
| | - Amel Ibrahim Othman
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | | | - Osama Azmy
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt; Department of Reproductive Health Research, Medical Research Division, National Research Centre, Cairo, Egypt; Egypt Center for Research and Regenerative Medicine, Cairo, Egypt
| |
Collapse
|
16
|
Amani S, Shahrooz R, Hobbenaghi R, Mohammadi R, Baradar Khoshfetrat A, Karimi A, Bakhtiari Z, Adcock IM, Mortaz E. Angiogenic effects of cell therapy within a biomaterial scaffold in a rat hind limb ischemia model. Sci Rep 2021; 11:20545. [PMID: 34654868 PMCID: PMC8519994 DOI: 10.1038/s41598-021-99579-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/21/2021] [Indexed: 11/26/2022] Open
Abstract
Critical limb ischemia (CLI) is a life- and limb-threatening condition affecting 1-10% of humans worldwide with peripheral arterial disease. Cellular therapies, such as bone marrow-derived mesenchymal stem cells (MSCs) have been used for the treatment of CLI. However, little information is available regarding the angiogenic potency of MSCs and mast cells (MC) in angiogenesis. The aim of this study was to evaluate the ability of MCs and MSCs to induce angiogenesis in a rat model of ischemic hind limb injury on a background of a tissue engineered hydrogel scaffold. Thirty rats were randomly divided into six control and experimental groups as follows: (a) Control healthy (b) Ischemic positive control with right femoral artery transection, (c) ischemia with hydrogel scaffold, (d) ischemia with hydrogel plus MSC, (e) ischemia with hydrogel plus MC and (f) ischemia with hydrogel plus MSC and MCs. 106 of each cell type, isolated from bone marrow stroma, was injected into the transected artery used to induce hind limb ischemia. The other hind limb served as a non-ischemic control. After 14 days, capillary density, vascular diameter, histomorphometry and immunohistochemistry at the transected location and in gastrocnemius muscles were evaluated. Capillary density and number of blood vessels in the region of the femoral artery transection in animals receiving MSCs and MCs was increased compared to control groups (P < 0.05). Generally the effect of MCs and MSCs was similar although the combined MC/MSC therapy resulted in a reduced, rather than enhanced, effect. In the gastrocnemius muscle, immunohistochemical and histomorphometric observation showed a great ratio of capillaries to muscle fibers in all the cell-receiving groups (P < 0.05). The data indicates that the combination of hydrogel and cell therapy generates a greater angiogenic potential at the ischemic site than cell therapy or hydrogels alone.
Collapse
Affiliation(s)
- Saeede Amani
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Shahrooz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Rahim Hobbenaghi
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rahim Mohammadi
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | | | - Ali Karimi
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Zahra Bakhtiari
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Ian M Adcock
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Bone Marrow Mesenchymal Stem Cell Condition Medium Loaded on PCL Nanofibrous Scaffold Promoted Nerve Regeneration After Sciatic Nerve Transection in Male Rats. Neurotox Res 2021; 39:1470-1486. [PMID: 34309780 DOI: 10.1007/s12640-021-00391-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/09/2023]
Abstract
Nowadays, researchers pay a vast deal of attention to neural tissue regeneration due to its tremendous effect on the patient's life. There are many strategies, from using conventional autologous nerve grafts to the newly developed methods for reconstructing damaged nerves. Among the various therapeutic methods, incorporating highly potent biomolecules and growth factors, the damaged nerve site would promote nerve regeneration. The aim was to examine the efficiency of a mesenchymal stem cell condition medium (MSC-CM) loaded on a 3D-polycaprolactone (PCL) scaffold as a nerve conduit in an axotomy rat model. Twenty-four mature male rats were classified into four groups: controls (the animals of this group were intact), axotomy (10 mm piece of the nerve was removed), axotomy (10-mm piece of the nerve was removed) + scaffold, and axotomy (10-mm piece of the nerve was removed) + MSC-CM-loaded scaffold. We followed up nerve motor function using a sciatic function index and electromyography activity of the gastrocnemius muscle. At 12 weeks post axotomy, sciatic nerve and dorsal root ganglion specimens and L4 and L5 spinal cord segments were separated from the rats and were analyzed by stereological, immunohistochemistry, and RT-PCR procedures. The rats of the axotomy group presented the expected gross locomotor deficit. Stereological parameters, immunohistochemistry of GFAP, and gene expression of S100, NGF, and BDNF were significantly enhanced in the CM-loaded scaffold group compared with the axotomy group. The most observed similarity was noted between the results of the control group and the CM-loaded scaffold group. Our results support the potential applicability of MSC-CM-loaded PCL nanofibrous scaffold to treat peripheral nerve injury (PNI).
Collapse
|
18
|
Sotoodehnejadnematalahi F, Moghadasali R, Hajinasrollah M, Ehsani E, Hajizadeh-Saffar E, Sodeifi N, Saidi R, Zarrabi M, Farzanehkhah M, Sadeghi B, Baharvand H, Aghdami N. Immunomodulatory Activity of Human Bone Marrow and Adipose-Derived Mesenchymal Stem Cells Prolongs Allogenic Skin Graft Survival in Nonhuman Primates. CELL JOURNAL 2021; 23:1-13. [PMID: 33650815 PMCID: PMC7944119 DOI: 10.22074/cellj.2021.6895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/09/2019] [Indexed: 11/04/2022]
Abstract
Objective In the present study, we examined the tolerance-inducing effects of human adipose-derived mesenchymal stem cells (hAD-MSCs) and bone marrow-derived MSCs (hBM-MSCs) on a nonhuman primate model of skin transplantation. Materials and Methods In this experimental study, allogenic and xenogeneic of immunomodulatory properties of human AD-MSCs and BM-MSCs were evaluated by mixed lymphocyte reaction (MLR) assays. Human MSCs were obtained from BM or AD tissues (from individuals of either sex with an age range of 35 to 65 years) and intravenously injected (2×106 MSCs/kg) after allogeneic skin grafting in a nonhuman primate model. The skin sections were evaluated by H and E staining for histopathological evaluations, particularly inflammation and rejection reaction of grafts after 96 hours of cell injection. At the mRNA and protein levels, cellular mediators of inflammation, such as CD4+IL-17+ (T helper 17; Th17) and CD4+INF-γ+ (T helper 1, Th1) cells, along with CD4+FoxP3+ cells (Treg), as the mediators of immunomodulation, were measured by RT-PCR and flow cytometry analyses. Results A significant Treg cells expansion was observed in MSCs-treated animals which reached the zenith at 24 hours and remained at a high concentration for 96 hours; however, Th1 and Th17 cells were significantly decreased. Our results showed that human MSCs significantly decrease Th1 and Th17 cell proliferation by decreasing interleukin-17 (IL-17) and interferon-γ (INF-γ) production and significantly increase Treg cell proliferation by increasing FoxP3 production. They also extend the allogenic skin graft survival in nonhuman primates. Histological evaluations showed no obvious presence of inflammatory cells or skin redness or even bulging after MSCs injection up to 96 hours, compared to the group without MSCs. There were no significant differences between hBM-MSCs and hAD-MSCs in terms of histopathological scores and inflammatory responses (P<0.05). Conclusion It seems that MSCs could be regarded as a valuable immunomodulatory tool to reduce the use of immunosuppressive agents.
Collapse
Affiliation(s)
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ehsan Ehsani
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Sodeifi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Reza Saidi
- Department of Surgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | | | - Bahareh Sadeghi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Hossein Baharvand
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
19
|
Influence of administration of mesenchymal stromal cell on pediatric oxygenator performance and inflammatory response. ACTA ACUST UNITED AC 2021; 5:99-107. [PMID: 33899029 PMCID: PMC8064572 DOI: 10.1016/j.xjon.2021.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective Mesenchymal stromal cells have important immunomodulatory and neuroprotective properties. The aim of this study was to evaluate the feasibility of mesenchymal stromal cell administration into a cardiopulmonary bypass (CPB) circuit, including a pediatric oxygenator, and to assess the immunomodulatory response of the circulating blood prime. Methods A bypass circuit with a pediatric oxygenator, including integral filter was primed with bank whole blood. Normal saline (control) or 120 × 106 mesenchymal stromal cells were injected into the venous reservoir after 80 minutes of perfusion. To assess oxygenator function, immune reaction, and cytokine/chemokine levels, the ex vivo circulation was maintained for 300 minutes after administration. Results There were no differences in flow rate, trans-oxygenator pressure gradient, blood oxygen, and carbon dioxide levels between control and cell delivery groups. No adhesion of mesenchymal stromal cells was observed on the filter mesh by scanning electron microscopy. Lymphocyte surface marker assay found no difference in the number of B cells, T cells, or natural killer cells between the 2 groups, indicating no immunogenicity of allogeneic mesenchymal stromal cells under ex vivo CPB conditions. CPB significantly changed the level of interleukin (IL) 4, IL-6, IL-8, IP-10, macrophage colony stimulating factor, macrophage inflammatory protein-1β, monocyte chemoattractant protein-1, and IL-1α over time. IL-6 level was significantly increased after cell administration. Conclusions The administration of mesenchymal stromal cells does not interfere with oxygenator function. Allogeneic mesenchymal stromal cells show no immunogenicity, and increase plasma IL-6 level during ex vivo circulation. Further investigation is necessary to determine the effect of mesenchymal stromal cell delivery through CPB during pediatric cardiac surgery.
Collapse
|
20
|
Biressi S, Filareto A, Rando TA. Stem cell therapy for muscular dystrophies. J Clin Invest 2021; 130:5652-5664. [PMID: 32946430 DOI: 10.1172/jci142031] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and.,Dulbecco Telethon Institute, University of Trento, Povo, Italy
| | - Antonio Filareto
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Conneticut, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
21
|
Rüger BM, Buchacher T, Dauber EM, Pasztorek M, Uhrin P, Fischer MB, Breuss JM, Leitner GC. De novo Vessel Formation Through Cross-Talk of Blood-Derived Cells and Mesenchymal Stromal Cells in the Absence of Pre-existing Vascular Structures. Front Bioeng Biotechnol 2020; 8:602210. [PMID: 33330432 PMCID: PMC7718010 DOI: 10.3389/fbioe.2020.602210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/26/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The generation of functional blood vessels remains a key challenge for regenerative medicine. Optimized in vitro culture set-ups mimicking the in vivo perivascular niche environment during tissue repair may provide information about the biological function and contribution of progenitor cells to postnatal vasculogenesis, thereby enhancing their therapeutic potential. AIM We established a fibrin-based xeno-free human 3D in vitro vascular niche model to study the interaction of mesenchymal stromal cells (MSC) with peripheral blood mononuclear cells (PBMC) including circulating progenitor cells in the absence of endothelial cells (EC), and to investigate the contribution of this cross-talk to neo-vessel formation. MATERIALS AND METHODS Bone marrow-derived MSC were co-cultured with whole PBMC, enriched monocytes (Mo), enriched T cells, and Mo together with T cells, respectively, obtained from leukocyte reduction chambers generated during the process of single-donor platelet apheresis. Cells were embedded in 3D fibrin matrices, using exclusively human-derived culture components without external growth factors. Cytokine secretion was analyzed in supernatants of 3D cultures by cytokine array, vascular endothelial growth factor (VEGF) secretion was quantified by ELISA. Cellular and structural re-arrangements were characterized by immunofluorescence and confocal laser-scanning microscopy of topographically intact 3D fibrin gels. RESULTS 3D co-cultures of MSC with PBMC, and enriched Mo together with enriched T cells, respectively, generated, within 2 weeks, complex CD31+/CD34+ vascular structures, surrounded by basement membrane collagen type-IV+ cells and matrix, in association with increased VEGF secretion. PBMC contained CD31+CD34+CD45dimCD14- progenitor-type cells, and EC of neo-vessels were PBMC-derived. Vascular structures showed intraluminal CD45+ cells that underwent apoptosis thereby creating a lumen. Cross-talk of MSC with enriched Mo provided a pro-angiogenic paracrine environment. MSC co-cultured with enriched T cells formed "cell-in-cell" structures generated through internalization of T cells by CD31+CD45 dim/ - cells. No vascular structures were detected in co-cultures of MSC with either Mo or T cells. CONCLUSION Our xeno-free 3D in vitro vascular niche model demonstrates that a complex synergistic network of cellular, extracellular and paracrine cross-talk can contribute to de novo vascular development through self-organization via co-operation of immune cells with blood-derived progenitor cells and MSC, and thereby may open a new perspective for advanced vascular tissue engineering in regenerative medicine.
Collapse
Affiliation(s)
- Beate M. Rüger
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eva-Maria Dauber
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Pasztorek
- Department of Health Sciences, Medicine and Research, Faculty of Health and Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Pavel Uhrin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael B. Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
- Department of Health Sciences, Medicine and Research, Faculty of Health and Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Johannes M. Breuss
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gerda C. Leitner
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Xiong Y, Wang Y, Zhang J, Zhao N, Zhang H, Zhang A, Zhao D, Yu Z, Yin Y, Song L, Xiong Y, Luan X. hPMSCs protects against D-galactose-induced oxidative damage of CD4 + T cells through activating Akt-mediated Nrf2 antioxidant signaling. Stem Cell Res Ther 2020; 11:468. [PMID: 33148324 PMCID: PMC7641865 DOI: 10.1186/s13287-020-01993-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/22/2020] [Indexed: 12/27/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) were considered a regenerative therapeutic approach in both acute and chronic diseases. However, whether MSCs regulate the antioxidant metabolism of CD4+ T cells and weaken immunosenescence remains unclear. Here, we reported the protective effects of hPMSCs in aging-related CD4+ T cell senescence and identified the underlying mechanisms using a d-gal-induced mouse aging model. Methods In vivo study, 40 male C57BL/6 mice (8 weeks) were randomly divided into four groups: control group, d-gal group, hPMSC group, and PBS group. In in vitro experiment, human naive CD4+ T (CD4CD45RA) cells were prepared using a naive CD4+ T cell isolation kit II and pretreated with the Akt inhibitor LY294002 and Nrf2 inhibitor ML385. Then, isolated naive CD4+ T cell were co-cultured with hPMSCs for 72 h in the absence or presence of anti-CD3/CD28 Dynabeads and IL-2 as a mitogenic stimulus. Intracellular ROS changes were detected by flow cytometry. The activities of the antioxidant enzymes superoxide dismutase, glutathione peroxidase, and catalase were measured by colorimetric analysis. The senescent T cells were detected SA-β-gal stain. The expression of aging-related proteins was detected by Western blotting, RT-PCR, and confocal microscopy. Results We found that hPMSC treatment markedly decreased the ROS level, SA-β-gal-positive cells number, senescence-associated secretory phenotype (IL-6 and OPN) expression, and aging-related protein (P16 and P21) expression in senescent CD4+ T cells. Furthermore, hPMSC treatment effectively upregulated Nrf2 nuclear translocation and the expression of downstream target genes (HO-1, CAT, GCLC, and NQO1) in senescent CD4+ T cells. Moreover, in vitro studies revealed that hPMSCs attenuated CD4+ T cell senescence by upregulating the Akt/GSK-3β/Fyn pathway to activate Nrf2 functions. Conversely, the antioxidant effects of hPMSCs were blocked by the Akt inhibitor LY294002 and Nrf2 inhibitor ML385 in senescent CD4+ T cells. Conclusions Our results indicate that hPMSCs attenuate d-gal-induced CD4+ T cell senescence by activating Nrf2-mediated antioxidant defenses and that upregulation of Nrf2 by hPMSCs is regulated via the Akt/GSK-3β/Fyn pathway.
Collapse
Affiliation(s)
- Yanlian Xiong
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Yueming Wang
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Jiashen Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Nannan Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Hengchao Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Aiping Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Dongmei Zhao
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Zhenhai Yu
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Yancun Yin
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Lele Song
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Yanlei Xiong
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Xiying Luan
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, People's Republic of China.
| |
Collapse
|
23
|
Fričová D, Korchak JA, Zubair AC. Challenges and translational considerations of mesenchymal stem/stromal cell therapy for Parkinson's disease. NPJ Regen Med 2020; 5:20. [PMID: 33298940 PMCID: PMC7641157 DOI: 10.1038/s41536-020-00106-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta and the presence of Lewy bodies, which gives rise to motor and non-motor symptoms. Unfortunately, current therapeutic strategies for PD merely treat the symptoms of the disease, only temporarily improve the patients' quality of life, and are not sufficient for completely alleviating the symptoms. Therefore, cell-based therapies have emerged as a novel promising therapeutic approach in PD treatment. Mesenchymal stem/stromal cells (MSCs) have arisen as a leading contender for cell sources due to their regenerative and immunomodulatory capabilities, limited ethical concerns, and low risk of tumor formation. Although several studies have shown that MSCs have the potential to mitigate the neurodegenerative pathology of PD, variabilities in preclinical and clinical trials have resulted in inconsistent therapeutic outcomes. In this review, we strive to highlight the sources of variability in studies using MSCs in PD therapy, including MSC sources, the use of autologous or allogenic MSCs, dose, delivery methods, patient factors, and measures of clinical outcome. Available evidence indicates that while the use of MSCs in PD has largely been promising, conditions need to be standardized so that studies can be effectively compared with one another and experimental designs can be improved upon, such that this body of science can continue to move forward.
Collapse
Affiliation(s)
- Dominika Fričová
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jennifer A Korchak
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Abba C Zubair
- Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
24
|
Ran J, Fei Y, Wang C, Ruan D, Hu Y, Zheng Z, Chen X, Yin Z, Tang C, Chen Y, Huang J, Shen L, Wu L, Heng BC, Pioletti D, Shen W, Ouyang H. An Off-the-Shelf Tissue Engineered Cartilage Composed of Optimally Sized Pellets of Cartilage Progenitor/Stem Cells. ACS Biomater Sci Eng 2020; 7:881-892. [PMID: 33715373 DOI: 10.1021/acsbiomaterials.9b01863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Articular cartilage focal lesion remains an intractable challenge in sports medicine, and autologous chondrocytes' implantation (ACI) is one of the most commonly utilized treatment modality for this ailment. However, the current ACI technique requires two surgical steps which increases patients' morbidity and incurs additional medical costs. In the present study, we developed a one-step cryopreserved off-the-shelf ACI tissue-engineered (TE) cartilage by seeding pellets of spheroidal cartilage stem/progenitor cells (CSPCs) on a silk scaffold. The pellets were developed through a hanging-drop method, and the incubation time of 1 day could efficiently produce spheroidal pellets without any adverse influence on the cell activity. The pellet size was also optimized. Under chondrogenic induction, pellets consisting of 40 000 CSPCs were found to exhibit the most abundant cartilage matrix deposition and the highest mRNA expression levels of SOX9, aggrecan, and COL2A1, as compared with pellets consisting of 10 000, 100 000, or 200 000 CSPCs. Scaffolds seeded with CSPCs pellets containing 40 000 cells could be preserved in liquid nitrogen with the viability, migration, and chondrogenic ability remaining unaffected for as long as 3 months. When implanted in a rat trochlear cartilage defect model for 3 months, the ready-to-use, cryopreserved TE cartilage yielded fully cartilage reconstruction, which was comparable with the uncryopreserved control. Hence, our study provided preliminary data that our off-the-shell TE cartilage with optimally sized CSPCs pellets seeded within silk scaffolds exhibited strong cartilage repair capacity, which provided a convenient and promising one-step surgical approach to ACI.
Collapse
Affiliation(s)
- Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Canlong Wang
- Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yejun Hu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zefeng Zheng
- Department of Orthopedic Surgery, The Children's Hospital, School of Medicine, Zhejiang University,3333 Binsheng Road, Hangzhou, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yangwu Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Lingfang Shen
- Air Force Health Care Center for Special Services, 15 Yanggongdi Road, Hangzhou 310000, China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Boon Chin Heng
- Peking University School of Stomatology, 5 Yiheyuan Road, Beijing, China
| | - Dominique Pioletti
- Laboratory of Biomechanical Orthopedics, EPFL, MED 3 2626 (Bâtiment MED), Station 9, Lausanne CH-1015, Switzerland
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
25
|
Cabezas J, Rojas D, Wong Y, Telleria F, Manriquez J, Mançanares ACF, Rodriguez-Alvarez LL, Castro FO. In vitro preconditioning of equine adipose mesenchymal stem cells with prostaglandin E 2, substance P and their combination changes the cellular protein secretomics and improves their immunomodulatory competence without compromising stemness. Vet Immunol Immunopathol 2020; 228:110100. [PMID: 32871408 DOI: 10.1016/j.vetimm.2020.110100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSC) are modern tools in regenerative therapies of humans and animals owed to their immunomodulatory properties, which are activated in a pro-inflammatory environment. Different preconditioning strategies had been devised to enhance the immunomodulatory properties of MSC. In this research, we evaluated the immunological attributes of equine adipose MSC (eAMSC) before and after preconditioning in vitro with prostaglandin E2 (PGE2), substance P (SP), their combination and IFNγ. PGE2/SP was the best combination to keep or enhance the mesodermal lineage differentiation of eAMSC. Alongside with this, preconditioning of eMSC with PGE2 and SP did not affect expression of stemness MSC surface phenotype: CD90+, CD44+, MHC class I+, MHC class II- and CD45-, assessed by cytometry. Both naïve and preconditioned eAMSC expressed genes related with immune properties, such as MHC-I, PTGES, IL6, IL1A, TNFα and IL8 assessed by qPCR. Only TNFα was under expressed in treated cells, while the other markers were either overexpressed or not changed. In no cases MHC-II expression was detected. The antiproliferative effect of preconditioned eAMSC exposed to activated peripheral blood mononuclear cells (PBMC) showed that SP treatment significantly inhibited proliferation of LPS stimulated PBMC. When eAMSC were stimulated with Poly I:C, all the treatments significantly inhibited proliferation of stimulated PBMC (p < 0.05). Direct contact (coculture) between the preconditioned eAMSC and PBMC, induced a shift of significantly more (CD4/CD25/FOXP3)+ T-regulatory PBMC than naïve eAMSC. In the experiments of this research, we investigated the secreted proteomic profile of naïve and preconditioned eAMSC, 42 up-regulated and 40 down-regulated proteins were found in the proteomic assay. Our proteomic data revealed profound changes in the secretory pattern of MSC exposed to different treatments, compared to naïve eAMSC as well as among treatments. In overall, compared to naïve cells, the protein profile of preconditioned cells resembled the mesenchymal-epithelial transition (MET). Here we showed that the combined use of PGE2 and SP provoked in overall the highest expression of anti-inflammatory markers as well as lead to an increased acquisition of a T-regulatory phenotype in preconditioned eAMSC without affecting their "stemness".
Collapse
Affiliation(s)
- J Cabezas
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - D Rojas
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Sciences, Department of Animal Pathology, Chile.
| | - Y Wong
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - F Telleria
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - J Manriquez
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - A C F Mançanares
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - L L Rodriguez-Alvarez
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| | - F O Castro
- Universidad de Concepción, Campus Chillan, Faculty of Veterinary Science, Department of Animal Science, Laboratorio de Biotecnología Animal, Chile.
| |
Collapse
|
26
|
Mesenchymal stem cell therapy for ischemic stroke: A look into treatment mechanism and therapeutic potential. J Neurol 2020; 268:4095-4107. [DOI: 10.1007/s00415-020-10138-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
|
27
|
Goodman SB, Lin T. Modifying MSC Phenotype to Facilitate Bone Healing: Biological Approaches. Front Bioeng Biotechnol 2020; 8:641. [PMID: 32671040 PMCID: PMC7328340 DOI: 10.3389/fbioe.2020.00641] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Healing of fractures and bone defects normally follows an orderly series of events including formation of a hematoma and an initial stage of inflammation, development of soft callus, formation of hard callus, and finally the stage of bone remodeling. In cases of severe musculoskeletal injury due to trauma, infection, irradiation and other adverse stimuli, deficient healing may lead to delayed or non-union; this results in a residual bone defect with instability, pain and loss of function. Modern methods of mechanical stabilization and autologous bone grafting are often successful in achieving fracture union and healing of bone defects; however, in some cases, this treatment is unsuccessful because of inadequate biological factors. Specifically, the systemic and local microenvironment may not be conducive to bone healing because of a loss of the progenitor cell population for bone and vascular lineage cells. Autologous bone grafting can provide the necessary scaffold, progenitor and differentiated lineage cells, and biological cues for bone reconstruction, however, autologous bone graft may be limited in quantity or quality. These unfavorable circumstances are magnified in systemic conditions with chronic inflammation, including obesity, diabetes, chronic renal disease, aging and others. Recently, strategies have been devised to both mitigate the necessity for, and complications from, open procedures for harvesting of autologous bone by using minimally invasive aspiration techniques and concentration of iliac crest bone cells, followed by local injection into the defect site. More elaborate strategies (not yet approved by the U.S. Food and Drug Administration-FDA) include isolation and expansion of subpopulations of the harvested cells, preconditioning of these cells or inserting specific genes to modulate or facilitate bone healing. We review the literature pertinent to the subject of modifying autologous harvested cells including MSCs to facilitate bone healing. Although many of these techniques and technologies are still in the preclinical stage and not yet approved for use in humans by the FDA, novel approaches to accelerate bone healing by modifying cells has great potential to mitigate the physical, economic and social burden of non-healing fractures and bone defects.
Collapse
Affiliation(s)
- Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| |
Collapse
|
28
|
Dong R, Bai Y, Dai J, Deng M, Zhao C, Tian Z, Zeng F, Liang W, Liu L, Dong S. Engineered scaffolds based on mesenchymal stem cells/preosteoclasts extracellular matrix promote bone regeneration. J Tissue Eng 2020; 11:2041731420926918. [PMID: 32551034 PMCID: PMC7278336 DOI: 10.1177/2041731420926918] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Recently, extracellular matrix-based tissue-engineered bone is a promising approach to repairing bone defects, and the seed cells are mostly mesenchymal stem cells. However, bone remodelling is a complex biological process, in which osteoclasts perform bone resorption and osteoblasts dominate bone formation. The interaction and coupling of these two kinds of cells is the key to bone repair. Therefore, the extracellular matrix secreted by the mesenchymal stem cells alone cannot mimic a complex bone regeneration microenvironment, and the addition of extracellular matrix by preosteoclasts may contribute as an effective strategy for bone regeneration. Here, we established the mesenchymal stem cell/preosteoclast extracellular matrix -based tissue-engineered bones and demonstrated that engineered-scaffolds based on mesenchymal stem cell/ preosteoclast extracellular matrix significantly enhanced osteogenesis in a 3 mm rat femur defect model compared with mesenchymal stem cell alone. The bioactive proteins released from the mesenchymal stem cell/ preosteoclast extracellular matrix based tissue-engineered bones also promoted the migration, adhesion, and osteogenic differentiation of mesenchymal stem cells in vitro. As for the mechanisms, the iTRAQ-labeled mass spectrometry was performed, and 608 differentially expressed proteins were found, including the IGFBP5 and CXCL12. Through in vitro studies, we proved that CXCL12 and IGFBP5 proteins, mainly released from the preosteoclasts, contributed to mesenchymal stem cells migration and osteogenic differentiation, respectively. Overall, our research, for the first time, introduce pre-osteoclast into the tissue engineering of bone and optimize the strategy of constructing extracellular matrix-based tissue-engineered bone using different cells to simulate the natural bone regeneration environment, which provides new sight for bone tissue engineering.
Collapse
Affiliation(s)
- Rui Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Yun Bai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Jingjin Dai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Moyuan Deng
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chunrong Zhao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Zhansong Tian
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Fanchun Zeng
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Wanyuan Liang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Lanyi Liu
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
29
|
Abstract
Stem cell therapy offers a breakthrough opportunity for the improvement of ischemic heart diseases. Numerous clinical trials and meta-analyses appear to confirm its positive but variable effects on heart function. Whereas these trials widely differed in design, cell type, source, and doses reinjected, cell injection route and timing, and type of cardiac disease, crucial key factors that may favour the success of cell therapy emerge from the review of their data. Various types of cell have been delivered. Injection of myoblasts does not improve heart function and is often responsible for severe ventricular arrythmia occurrence. Using bone marrow mononuclear cells is a misconception, as they are not stem cells but mainly a mix of various cells of hematopoietic lineages and stromal cells, only containing very low numbers of cells that have stem cell-like features; this likely explain the neutral results or at best the modest improvement in heart function reported after their injection. The true existence of cardiac stem cells now appears to be highly discredited, at least in adults. Mesenchymal stem cells do not repair the damaged myocardial tissue but attenuate post-infarction remodelling and contribute to revascularization of the hibernated zone surrounding the scar. CD34+ stem cells - likely issued from pluripotent very small embryonic-like (VSEL) stem cells - emerge as the most convincing cell type, inducing structural and functional repair of the ischemic myocardial area, providing they can be delivered in large amounts via intra-myocardial rather than intra-coronary injection, and preferentially after myocardial infarct rather than chronic heart failure.
Collapse
Affiliation(s)
- Philippe Hénon
- CellProthera SAS and Institut de Recherche en Hématologie et Transplantation, CellProthera SAS 12 rue du Parc, 68100, Mulhouse, France.
| |
Collapse
|
30
|
Tavakoli S, Ghaderi Jafarbeigloo HR, Shariati A, Jahangiryan A, Jadidi F, Jadidi Kouhbanani MA, Hassanzadeh A, Zamani M, Javidi K, Naimi A. Mesenchymal stromal cells; a new horizon in regenerative medicine. J Cell Physiol 2020; 235:9185-9210. [PMID: 32452052 DOI: 10.1002/jcp.29803] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022]
Abstract
In recent decades, mesenchymal stromal cells (MSCs) biomedical utilizing has attracted worldwide growing attention. After the first report of the human MSCs obtaining from the bone marrow (BM) tissue, these cells were isolated from wide types of the other tissues, ranging from adipose tissue to dental pulp. Their specific characteristics, comprising self-renewality, multipotency, and availability accompanied by their immunomodulatory properties and little ethical concern denote their importance in the context of regenerative medicine. Considering preclinical studies, MSCs can modify immune reactions during tissue repair and restoration, providing suitable milieu for tissue recovery; on the other hand, they can be differentiated into comprehensive types of the body cells, such as osteoblast, chondrocyte, hepatocyte, cardiomyocyte, fibroblast, and neural cells. Though a large number of studies have investigated MSCs capacities in regenerative medicine in varied animal models, the oncogenic capability of unregulated MSCs differentiation must be more assessed to enable their application in the clinic. In the current review, we provide a brief overview of MSCs sources, isolation, and expansion as well as immunomodulatory activities. More important, we try to collect and discuss recent preclinical and clinical research and evaluate current challenges in the context of the MSC-based cell therapy for regenerative medicine.
Collapse
Affiliation(s)
- Shirin Tavakoli
- Department of Toxicology and Pharmacology, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Shariati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Jahangiryan
- Immunology Department, Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine (IBTO), Tehran, Iran
| | - Faezeh Jadidi
- Student Research Committee, Zarand School of Nursing, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammd Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Kamran Javidi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
31
|
Chang SH, Kim HJ, Park CG. Allogeneic ADSCs Induce the Production of Alloreactive Memory-CD8 T Cells through HLA-ABC Antigens. Cells 2020; 9:cells9051246. [PMID: 32443511 PMCID: PMC7290988 DOI: 10.3390/cells9051246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
We investigated the immunogenicity of allogeneic human adipose-derived mesenchymal stem cells (ADSCs) through the production of alloreactive-CD8 T and -memory CD8 T cells, based on their human leukocyte antigen (HLA) expression. In surface antigen analysis, ADSCs do not express co-stimulatory molecules, but expresses HLA-ABC, which is further increased by exposure to the pro-inflammatory cytokines as well as IFN-γ alone. For immunogenicity analysis, allogeneic ADSCs cultured in xenofree medium (XF-ADSCs) were incubated with the recipient immune cells for allogeneic-antigen stimulation. As a result, XF-ADSCs induced IFN-γ and IL-17A release by alloreactive-CD8 T cells and the production of alloreactive-CD8 T cell through a direct pathway, although they have immunomodulatory activity. In the analysis of alloreactive memory CD8 T cells, XF-ADSCs also significantly induced the production of CFSE-low-CD8 TEM and -CD8 TCM cells. However, HLA-blocking antibodies significantly inhibited the production of CFSE-low memory-CD8 T cells, indicating that HLAs are the main antigens responsible for the development of allogeneic ADSCs' immunogenicity. These results suggested that HLA surface antigens expressed in allogeneic MSCs should be solved in order to address concerns related to the immunogenicity problem.
Collapse
Affiliation(s)
- Sung-Ho Chang
- Departments of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea;
| | - Hyun Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Dermatology, Samsung Medical Center, Seoul 06351, Korea
| | - Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea;
- Institute of Endemic Diseases, Medical Research center, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-2-740-8308
| |
Collapse
|
32
|
Cheng J, Zhao ZW, Wen JR, Wang L, Huang LW, Yang YL, Zhao FN, Xiao JY, Fang F, Wu J, Miao YL. Status, challenges, and future prospects of stem cell therapy in pelvic floor disorders. World J Clin Cases 2020; 8:1400-1413. [PMID: 32368533 PMCID: PMC7190946 DOI: 10.12998/wjcc.v8.i8.1400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/30/2020] [Accepted: 04/08/2020] [Indexed: 02/05/2023] Open
Abstract
Pelvic floor disorders (PFDs) represent a group of common and frequently-occurring diseases that seriously affect the life quality of women, generally including stress urinary incontinence and pelvic organ prolapse. Surgery has been used as a treatment for PFD, but almost 30% of patients require subsequent surgery due to a high incidence of postoperative complications and high recurrence rates. Therefore, investigations of new therapeutic strategies are urgently needed. Stem cells possess strong multi-differentiation, self-renewal, immunomodulation, and angiogenesis abilities and they are able to differentiate into various cell types of pelvic floor tissues and thus provide a potential therapeutic approach for PFD. Recently, various studies using different autologous stem cells have achieved promising results by improving the pelvic ligament and muscle regeneration and conferring the tissue elasticity and strength to the damaged tissue in PFD, as well as reduced inflammatory reactions, collagen deposition, and foreign body reaction. However, with relatively high rates of complications such as bladder stone formation and wound infections, further studies are necessary to investigate the role of stem cells as maintainers of tissue homeostasis and modulators in early interventions including therapies using new stem cell sources, exosomes, and tissue-engineering combined with stem cell-based implants, among others. This review describes the types of stem cells and the possible interaction mechanisms in PFD treatment, with the hope of providing more promising stem cell treatment strategies for PFD in the future.
Collapse
Affiliation(s)
- Juan Cheng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, West China Campus, Sichuan University, Chengdu 610041, Sichuan Province, China
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zhi-Wei Zhao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ji-Rui Wen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ling Wang
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Wei Huang
- West China School of Stomatology Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan-Lin Yang
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Feng-Nian Zhao
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jing-Yue Xiao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Fei Fang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jiang Wu
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ya-Li Miao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, West China Campus, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
33
|
Weng T, Wu P, Zhang W, Zheng Y, Li Q, Jin R, Chen H, You C, Guo S, Han C, Wang X. Regeneration of skin appendages and nerves: current status and further challenges. J Transl Med 2020; 18:53. [PMID: 32014004 PMCID: PMC6996190 DOI: 10.1186/s12967-020-02248-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Tissue-engineered skin (TES), as an analogue of native skin, is promising for wound repair and regeneration. However, a major drawback of TES products is a lack of skin appendages and nerves to enhance skin healing, structural integrity and skin vitality. Skin appendages and nerves are important constituents for fully functional skin. To date, many studies have yielded remarkable results in the field of skin appendages reconstruction and nerve regeneration. However, patients often complain about a loss of skin sensation and even cutaneous chronic pain. Restoration of pain, temperature, and touch perceptions should now be a major challenge to solve in order to improve patients’ quality of life. Current strategies to create skin appendages and sensory nerve regeneration are mainly based on different types of seeding cells, scaffold materials, bioactive factors and involved signaling pathways. This article provides a comprehensive overview of different strategies for, and advances in, skin appendages and sensory nerve regeneration, which is an important issue in the field of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Pan Wu
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Wei Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Yurong Zheng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qiong Li
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Haojiao Chen
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Chuangang You
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Songxue Guo
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
34
|
Hsu MF, Yu SH, Chuang SJ, Kuo TKC, Singal PK, Huang CY, Kao CL, Kuo CH. Can mesenchymal stem cell lysate reverse aging? Aging (Albany NY) 2019; 10:2900-2910. [PMID: 30362957 PMCID: PMC6224235 DOI: 10.18632/aging.101595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022]
Abstract
Recent findings regarding uses of adipose-derived mesenchymal stem cell (MSC)-lysate on weight loss and improved glucose tolerance in mice on a high-fat diet suggest an encouraging possibility of using MSC lysate for an anti-aging intervention in humans. However, weight loss and lipopenia during late life can be as life-threatening as hyperglycemia during early adulthood. For this 3-year lifelong experiment, a total of 92 rats were randomized into the vehicle-injected group (F=22; M=24) and the MSC lysate injected group (F=22, M=24). We examined longevity, spontaneous locomotor activity, and body composition in rats maintained on a normal diet and received an intermittent treatment of human adipose-derived MSC lysate (3 times a week, 11 times a month given every second month), starting at 12 months of age until natural death. In substantiating previous knowledge regarding the effects of long-term MSC lysate treatments on fat loss and insulin resistance, the present findings also highlighted a shortened average lifespan, a longer inactive time, and a greater bone loss with a relative increase of lean mass in MSC lysate rats with respect to controls. Conclusion: Our data suggest that MSC lysate treatments stimulate disparity in tissue development and produce a cachexia-like effect to decrease longevity.
Collapse
Affiliation(s)
- Ming-Fen Hsu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Szu-Hsien Yu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Sheng-Ju Chuang
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,Université Catholique de Louvain and de Duve Institute, Brussels, Belgium
| | - Tom Kwang-Chun Kuo
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre and Department of Physiology and Pathophysiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Chih-Yang Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chung-Lan Kao
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital and National Yang Ming University, Taipei, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| |
Collapse
|
35
|
Pedrosa M, Gomes J, Laranjeira P, Duarte C, Pedreiro S, Antunes B, Ribeiro T, Santos F, Martinho A, Fardilha M, Domingues MR, Abecasis M, P da Silva JA, Paiva A. Immunomodulatory effect of human bone marrow-derived mesenchymal stromal/stem cells on peripheral blood T cells from rheumatoid arthritis patients. J Tissue Eng Regen Med 2019; 14:16-28. [PMID: 31502378 DOI: 10.1002/term.2958] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/04/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a Th1/Th17-mediated autoimmune disease whose current treatment, consisting in the blockage of inflammatory cytokines by disease-modifying antirheumatic drugs, is not effective for all patients. The therapeutic potential of mesenchymal stromal/stem cells' (MSCs) immunomodulatory properties is being explored in RA. Here, we investigate the effect of human bone marrow (BM)-MSCs on the expression of cytokines involved in RA physiopathology by the distinct functional compartments of CD4+ and CD8+ T cells from RA patients. Peripheral blood mononuclear cells from healthy individuals (n = 6) and RA patients (n = 12) were stimulated with phorbol myristate acetate plus ionomycin and cultured in the presence/absence of BM-MSCs. The expression of (interleukin) IL-2, tumor necrosis factor alpha (TNF-α), and interferon-gamma (IFN-γ) was evaluated in naive, central memory, effector memory, and effector CD4+ and CD8+ T cells, whereas IL-6, IL-9, and IL-17 expression was measured in total CD4+ and CD8+ T cells. mRNA expression of IL-4, IL-10, transforming growth factor beta (TGF-β), cytotoxic T-lymphocyte-associated antigen 4, and/or forkhead box P3 was quantified in fluorescence-activated cell sorting-purified CD4+ T cells, CD8+ T cells, and CD4+ Treg. BM-MSCs inhibited the production of TNF-α, IL-17, IL-6, IL-2, IFN-γ, and IL-9 by T cells from RA patients, mainly by reducing the percentage of cells producing cytokines. This inhibitory effect was transversal to all T cell subsets analyzed. At mRNA level, BM-MSCs increased expression of IL-10 and TGF-β by CD4+ and CD8+ T cells. BM-MSCs displayed a striking inhibitory action over T cells from RA patients, reducing the expression of cytokines involved in RA physiopathology. Remarkably, BM-MSC-derived immunomodulation affected either naive, effector, and memory T cells.
Collapse
Affiliation(s)
- Mónia Pedrosa
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal
| | - Joana Gomes
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Paula Laranjeira
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Cátia Duarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Susana Pedreiro
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | | | - Tânia Ribeiro
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal
| | - Francisco Santos
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal.,Stemlab SA, Cantanhede, Portugal
| | - António Martinho
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | - Margarida Fardilha
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal.,Laboratory of Signal Transduction, Institute of Biomedicine-iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,Departament of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Manuel Abecasis
- Serviço de Transplantação de Progenitores Hematopoiéticos (UTM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - José António P da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Artur Paiva
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Instituto Politecnico de Coimbra, ESTESC-Coimbra Health School, Ciencias Biomedicas Laboratoriais, Coimbra, Portugal
| |
Collapse
|
36
|
Maeda T, Sarkislali K, Leonetti C, Kapani N, Dhari Z, Al Haj I, Ulrey R, Hanley PJ, Jonas RA, Ishibashi N. Impact of Mesenchymal Stromal Cell Delivery Through Cardiopulmonary Bypass on Postnatal Neurogenesis. Ann Thorac Surg 2019; 109:1274-1281. [PMID: 31563487 DOI: 10.1016/j.athoracsur.2019.08.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Neurodevelopmental impairment is an important challenge for survivors after neonatal surgery with cardiopulmonary bypass (CPB). The subventricular zone, where most neural stem/progenitors originate, plays a critical role in cortical maturation of the frontal lobe. Promoting neurogenesis in the subventricular zone is therefore a potential therapeutic target for preserving cortical growth. Mesenchymal stromal cells (MSCs) promote endogenous regeneration in the rodent brain. We investigated the impact of MSC delivery through CPB on neural stem/progenitor cells and neuroblasts (ie, young neurons) in the piglet subventricular zone. METHODS Two-week-old piglets (n = 12) were randomly assigned to one of three groups: (1) control, (2) deep hypothermic circulatory arrest, and (3) circulatory arrest, followed by MSC administration. MSCs (10 × 106 per kg) were delivered through CPB during the rewarming period. Neural stem/progenitors, proliferating cells, and neuroblasts were identified with immunohistochemistry at 3 hours after CPB. RESULTS CPB-induced insults caused an increased proliferation of neural stem/progenitors (P < .05). MSC delivery reduced the acute proliferation. MSC treatment increased the number of neuroblasts in the outer region of the subventricular zone (P < .05) where they form migrating chains toward the frontal lobe. Conversely, the thickness of the neuroblast-dense band along the lateral ventricle was reduced after treatment (P < .05). These findings suggest that MSC treatment changes neuroblast distribution within the subventricular zone. CONCLUSIONS MSC delivery through CPB has the potential to mitigate effects of CPB on neural stem/progenitor cells and to promote migration of neuroblasts. Further investigation is necessary to determine the long-term effect of MSC treatment during CPB on postnatal neurogenesis.
Collapse
Affiliation(s)
- Takuya Maeda
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC
| | - Kamil Sarkislali
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC
| | - Camille Leonetti
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC
| | - Nisha Kapani
- George Washington University School of Medicine and Health Science, Washington, DC
| | - Zaenab Dhari
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC
| | - Ibtisam Al Haj
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC
| | - Robert Ulrey
- Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation and Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC
| | - Patrick J Hanley
- George Washington University School of Medicine and Health Science, Washington, DC; Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation and Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC
| | - Richard A Jonas
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; George Washington University School of Medicine and Health Science, Washington, DC
| | - Nobuyuki Ishibashi
- Children's National Heart Institute, Children's National Hospital, Washington, DC; Center for Neuroscience Research, Children's National Hospital, Washington, DC; George Washington University School of Medicine and Health Science, Washington, DC.
| |
Collapse
|
37
|
Improvement in viability and mineralization of osteoporotic bone marrow mesenchymal stem cell through combined application of photobiomodulation therapy and oxytocin. Lasers Med Sci 2019; 35:557-566. [DOI: 10.1007/s10103-019-02848-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022]
|
38
|
Hoogduijn MJ, Lombardo E. Mesenchymal Stromal Cells Anno 2019: Dawn of the Therapeutic Era? Concise Review. Stem Cells Transl Med 2019; 8:1126-1134. [PMID: 31282113 PMCID: PMC6811696 DOI: 10.1002/sctm.19-0073] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
2018 was the year of the first marketing authorization of an allogeneic stem cell therapy by the European Medicines Agency. The authorization concerns the use of allogeneic adipose tissue-derived mesenchymal stromal cells (MSCs) for treatment of complex perianal fistulas in Crohn's disease. This is a breakthrough in the field of MSC therapy. The last few years have, furthermore, seen some breakthroughs in the investigations into the mechanisms of action of MSC therapy. Although the therapeutic effects of MSCs have largely been attributed to their secretion of immunomodulatory and regenerative factors, it has now become clear that some of the effects are mediated through host phagocytic cells that clear administered MSCs and in the process adapt an immunoregulatory and regeneration supporting function. The increased interest in therapeutic use of MSCs and the ongoing elucidation of the mechanisms of action of MSCs are promising indicators that 2019 may be the dawn of the therapeutic era of MSCs and that there will be revived interest in research to more efficient, practical, and sustainable MSC-based therapies. Stem Cells Translational Medicine 2019;8:1126-1134.
Collapse
Affiliation(s)
- Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
39
|
Riazifar M, Mohammadi MR, Pone EJ, Yeri A, Lässer C, Segaliny AI, McIntyre LL, Shelke GV, Hutchins E, Hamamoto A, Calle EN, Crescitelli R, Liao W, Pham V, Yin Y, Jayaraman J, Lakey JRT, Walsh CM, Van Keuren-Jensen K, Lotvall J, Zhao W. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS NANO 2019; 13:6670-6688. [PMID: 31117376 PMCID: PMC6880946 DOI: 10.1021/acsnano.9b01004] [Citation(s) in RCA: 357] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
To dissect therapeutic mechanisms of transplanted stem cells and develop exosome-based nanotherapeutics in treating autoimmune and neurodegenerative diseases, we assessed the effect of exosomes secreted from human mesenchymal stem cells (MSCs) in treating multiple sclerosis using an experimental autoimmune encephalomyelitis (EAE) mouse model. We found that intravenous administration of exosomes produced by MSCs stimulated by IFNγ (IFNγ-Exo) (i) reduced the mean clinical score of EAE mice compared to PBS control, (ii) reduced demyelination, (iii) decreased neuroinflammation, and (iv) upregulated the number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords of EAE mice. Co-culture of IFNγ-Exo with activated peripheral blood mononuclear cells (PBMCs) cells in vitro reduced PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines including IL-6, IL-12p70, IL-17AF, and IL-22 yet increased levels of immunosuppressive cytokine indoleamine 2,3-dioxygenase. IFNγ-Exo could also induce Tregs in vitro in a murine splenocyte culture, likely mediated by a third-party accessory cell type. Further, IFNγ-Exo characterization by deep RNA sequencing suggested that IFNγ-Exo contains anti-inflammatory RNAs, where their inactivation partially hindered the exosomes potential to induce Tregs. Furthermore, we found that IFNγ-Exo harbors multiple anti-inflammatory and neuroprotective proteins. These results not only shed light on stem cell therapeutic mechanisms but also provide evidence that MSC-derived exosomes can potentially serve as cell-free therapies in creating a tolerogenic immune response to treat autoimmune and central nervous system disorders.
Collapse
Affiliation(s)
- Milad Riazifar
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - M. Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Egest J. Pone
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Department of Physiology and Biophysics, Vaccine Research and Development Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ashish Yeri
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Cecilia Lässer
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Aude I. Segaliny
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Laura L. McIntyre
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ganesh Vilas Shelke
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg 41345, Sweden
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Ashley Hamamoto
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Erika N. Calle
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Rossella Crescitelli
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Wenbin Liao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Victor Pham
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Yanan Yin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jayapriya Jayaraman
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California, Irvine, Orange, California 92868, United States
| | - Craig M. Walsh
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Kendall Van Keuren-Jensen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Jan Lotvall
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Weian Zhao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Corresponding Author:
| |
Collapse
|
40
|
Murphy N, Treacy O, Lynch K, Morcos M, Lohan P, Howard L, Fahy G, Griffin MD, Ryan AE, Ritter T. TNF-α/IL-1β-licensed mesenchymal stromal cells promote corneal allograft survival via myeloid cell-mediated induction of Foxp3 + regulatory T cells in the lung. FASEB J 2019; 33:9404-9421. [PMID: 31108041 DOI: 10.1096/fj.201900047r] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stromal cells (MSCs) have shown promise as a therapy for immune-mediated disorders, including transplant rejection. Our group previously demonstrated the efficacy of pretransplant, systemic administration of allogeneic but not syngeneic MSCs in a rat cornea transplant model. The aim of this study was to enhance the immunomodulatory capacity of syngeneic MSCs. In vitro, MSCs licensed with TNF-α/IL-1β (MSCsTNF-α/IL-1β) suppress syngeneic lymphocyte proliferation via NO production. In vivo, when administered post-transplantation, nonlicensed syngeneic MSCs improved graft survival from 0 to 50% and MSCsTNF-α/IL-1β, in an NO-dependent manner, improved survival to 70%. Improved survival was associated with increased CD4+CD25+forkhead box P3+ regulatory T (Treg) cells and decreased proinflammatory cytokine expression in the draining lymph node. MSCsTNF-α/IL-1β demonstrated a more potent immunomodulatory capacity compared with nonlicensed MSCs, promoting an immune-regulatory CD11b+B220+ monocyte/macrophage population and significantly expanding Treg cells in the lungs and spleen. Ex vivo, we observed that lung-derived myeloid cells act as intermediaries of MSC immunomodulatory function. MSC-conditioned myeloid cells suppressed stimulated lymphocyte proliferation and promoted expansion of Treg cells from naive lymphocytes. This work illustrates how syngeneic MSC therapy can be enhanced by licensing and optimization of timing strategies and further highlights the important role of myeloid cells in mediating MSC immunomodulatory capacity.-Murphy, N., Treacy, O., Lynch, K., Morcos, M., Lohan, P., Howard, L., Fahy, G., Griffin, M. D., Ryan, A. E., Ritter, T. TNF-α/IL-1β-licensed mesenchymal stromal cells promote corneal allograft survival via myeloid cell-mediated induction of Foxp3+ regulatory T cells in the lung.
Collapse
Affiliation(s)
- Nick Murphy
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Oliver Treacy
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Kevin Lynch
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Maurice Morcos
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Paul Lohan
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Linda Howard
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Gerry Fahy
- Department of Ophthalmology, University Hospital Galway, National University of Ireland-Galway, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Aideen E Ryan
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), School of Medicine, College of Medicine, Nursing, and Health Sciences, National University of Ireland-Galway, Galway, Ireland
| |
Collapse
|
41
|
Calonge M, Pérez I, Galindo S, Nieto-Miguel T, López-Paniagua M, Fernández I, Alberca M, García-Sancho J, Sánchez A, Herreras JM. A proof-of-concept clinical trial using mesenchymal stem cells for the treatment of corneal epithelial stem cell deficiency. Transl Res 2019; 206:18-40. [PMID: 30578758 DOI: 10.1016/j.trsl.2018.11.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/18/2018] [Accepted: 11/18/2018] [Indexed: 01/02/2023]
Abstract
Ocular stem cell transplantation derived from either autologous or allogeneic donor corneoscleral junction is a functional cell therapy to manage extensive and/or severe limbal stem cell deficiencies that lead to corneal epithelial failure. Mesenchymal stem cells have been properly tested in animal models of this ophthalmic pathology, but never in human eyes despite their potential advantages. We conducted a 6- to 12-month proof-of-concept, randomized, and double-masked pilot trial to test whether allogeneic bone marrow-derived mesenchymal stem cell transplantation (MSCT], n = 17) was as safe and as equally efficient as allogeneic cultivated limbal epithelial transplantation (CLET), (n = 11) to improve corneal epithelial damage due to limbal stem cell deficiency. Primary endpoints demanded combination of symptoms, signs, and the objective improvement of the epithelial phenotype in central cornea by in vivo confocal microscopy. This proof-of-concept trial showed that MSCT was as safe and efficacious as CLET. Global success at 6-12 months was 72.7%-77.8% for CLET cases and 76.5%-85.7% for MSCT cases (not significant differences). Central corneal epithelial phenotype improved in 71.4% and 66.7% of MSCT and CLET cases, respectively at 12 months (P = 1.000). There were no adverse events related to cell products. This trial suggests first evidence that MSCT facilitated improvement of a diseased corneal epithelium due to lack of its stem cells as efficiently as CLET. Consequently, not only CLET but also MSCT deserves more preclinical investigational resources before the favorable results of this proof-of-concept trial could be transformed into the larger numbers of the multicenter trials that would provide stronger evidence. (ClinicalTrials.gov number, NCT01562002.).
Collapse
Affiliation(s)
- Margarita Calonge
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| | - Inmaculada Pérez
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain.
| | - Sara Galindo
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| | - Teresa Nieto-Miguel
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| | - Marina López-Paniagua
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| | - Itziar Fernández
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| | - Mercedes Alberca
- IBGM (Institute of Molecular Biology and Genetics), University of Valladolid and National Research Council (CSIC), and University Scientific Park, Valladolid, Spain.
| | - Javier García-Sancho
- IBGM (Institute of Molecular Biology and Genetics), University of Valladolid and National Research Council (CSIC), and University Scientific Park, Valladolid, Spain.
| | - Ana Sánchez
- IBGM (Institute of Molecular Biology and Genetics), University of Valladolid and National Research Council (CSIC), and University Scientific Park, Valladolid, Spain.
| | - José M Herreras
- IOBA (Institute of Applied Ophthalmobiology), University of Valladolid, Valladolid, Spain; CIBER-BBN (Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain.
| |
Collapse
|
42
|
Allogeneic ADSCs induce CD8 T cell-mediated cytotoxicity and faster cell death after exposure to xenogeneic serum or proinflammatory cytokines. Exp Mol Med 2019; 51:1-10. [PMID: 30858365 PMCID: PMC6412000 DOI: 10.1038/s12276-019-0231-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/02/2018] [Accepted: 10/29/2018] [Indexed: 12/28/2022] Open
Abstract
This study examined the induction of recipient T-cell cytotoxicity after exposure to allogeneic adipose-derived mesenchymal stem cells (ADSCs). ADSCs pre-exposed to xenogeneic serum significantly induced cytotoxicity through CD8 T-cell granzyme B secretion after allogeneic antigen stimulation, and this effect was increased with prolonged reaction time. ADSCs pretreated with proinflammatory cytokines also induced cytotoxicity through granzyme B secretion and significantly increased human leukocyte antigen (HLA)-ABC expression. T-cell cytotoxicity towards ADSCs grown in xeno-free medium (XF-ADSCs) was lower than that towards ADSCs exposed to xenogeneic serum or proinflammatory cytokines, but XF-ADSCs still induced cytotoxicity. We further investigated the causes of T-cell cytotoxicity towards XF-ADSCs. XF-ADSC death was effectively inhibited by HLA-blocking antibodies, suggesting that ADSC HLAs are a major cause of alloreactive T-cell generation. These results indicated that culturing of allogeneic ADSCs with recipient serum may alleviate alloreactive CD8 T-cell cytotoxicity. Ultimately, development of therapeutic agents using autologous ADSCs would be a suitable way to avoid immunogenicity and CD8 T cell-mediated cytotoxicity, but more attention should be paid to the potential immunogenicity of allogeneic ADSCs, which could perhaps be mitigated through the use of immunosuppressants. The conditions under which donor stem cells are cultured can limit attack by the recipient’s immune system after transplantation. Adult stem cells taken from donors who are genetically similar to recipients show promise as therapeutic agents for various conditions, from cardiac to immunity-related diseases. However, patients’ immune systems will often attack and destroy the transplanted cells. Chung-Gyu Park and Sung-Ho Chang at Seoul National University, South Korea, explored methods of growing stem cells so that they are less likely to be destroyed by the patient’s T-cells. The team found that human T-cells will attack stem cells grown in media containing bovine serum or those pre-treated with pro-inflammatory proteins. T-cell activity was weaker against stem cells grown in media with autologous serum.
Collapse
|
43
|
Ge Y, Zhang Y, Tang Q, Gao J, Yang H, Gao Z, Zhao RC. Mechanisms of the Immunomodulation Effects of Bone Marrow-Derived Mesenchymal Stem Cells on Facial Nerve Injury in Sprague-Dawley Rats. Stem Cells Dev 2019; 28:489-496. [PMID: 30704338 DOI: 10.1089/scd.2018.0104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Normal facial nerve (FN) function is very important for human being. However, if injured, FN function is difficult to restore completely. Recently, many studies reported the immune regulation function of stem cells (SCs). However, the immunomodulation function of SCs on FN injury is still unclear. Our study aims to explore the mechanism of immunomodulation effect of Sprague-Dawley rat bone marrow-derived SCs (BMSCs) on FN injury and specially focus on the regulation of Th17 and the protection effects of BMSCs on central facial motor neurons (FMNs). First, rat FNs were harvested. FN and BMSCs were cultured together or separately and levels of transforming growth factor (TGF)-β1, interleukin (IL)-6, hepatocyte growth factor (HGF), inducible nitric oxide synthase (iNOS), and prostaglandin E2 (PGE2) in supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Then, after treating with or without local BMSCs injection, the proportion of Th17 in neck lymph nodes (LNs) was investigated in rat FN injury models. Furthermore, the apoptotic index of FMNs was studied in rat FN injury models that were treated with or without BMSCs. We found that BMSCs could secrete high levels of IL-6, HGF, PGE2, iNOS, and TGF-β1 in culture. The percentage of Th17 of neck LNs in BMSCs-treated group was significantly lower than that in the control group. The apoptotic index of FMNs in BMSCs-treated group was significantly lower than that in the control group. In conclusion, our research indicates BMSCs could independently secrete cytokines IL-6, HGF, PGE2, iNOS, and TGF-β1, and these cytokines could regulate the balance among subsets of CD4+ T cells and could protect FMNs by inhibiting neuron apoptosis.
Collapse
Affiliation(s)
- Yining Ge
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,3 Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongli Zhang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Tang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juanjuan Gao
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Yang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Gao
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- 4 Department of Cell Biology, Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Monteiro BS, Santos BSD, Almeida BLD, Hiura E, Fiorio WAB, Valdetaro GP, Gonçalves DV, Silva CS, Champion T, Campagnol D. Adipose tissue derived mesenchymal stem cell transplantation in the treatment of ischemia/reperfusion induced acute kidney injury in rats. Application route and therapeutic window1. Acta Cir Bras 2019; 33:1016-1026. [PMID: 30517328 DOI: 10.1590/s0102-865020180110000008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/25/2018] [Indexed: 01/23/2023] Open
Abstract
PURPOSE To evaluate renal repair in rats who had renal infarction induced by the obstruction of blood flow in the renal artery and were treated with transplantation of adipose tissue derived mesenchymal stem cell. METHODS 16-week-old Wistar rats (n=72) were used, submitted to celiotomy and had of the renal artery and vein clipped for 24 hours. The animals were randomly assigned to 10 experimental homogeneous groups, corresponding to the treatments with phosphate-buffered saline (PBS) or adipose tissue derived mesenchymal stem cell (ADSC), duration of application (24 or 48 hours), and site of transplantation (lateral vein of the tail or intrarenal). After the treatments were performed, at 8 and 31 days, four animals in each group were subjected to left nephrectomy for histological studies. RESULTS Histologically, a higher amount of cell debris and tubules devoid of the epithelium and a higher degree of necrosis were observed in the groups treated with PBS, as opposed to a low degree of necrosis and higher tubular vascularization in the groups treated with ADSC, particularly in the group treated with intrarenal ADSC 48 hours after injury. CONCLUSION The transplantation of ADSC positively contributed to the replacement of necrotic tissue by renal tubular cells, vascularization of the renal parenchyma, and restoration of the organ function.
Collapse
Affiliation(s)
- Betânia Souza Monteiro
- Full Professor, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, Universidade Vila Velha (UVV), Brazil. Conception and design of the study, manuscript writing, critical revision, final approval
| | - Bianka Souza Dos Santos
- Veterinary Medical, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, UVV, Vila Velha-ES, Brazil. Technical procedures, acquisition of data, manuscript preparation and writing
| | - Bruna Lopes de Almeida
- Veterinary Medical, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, UVV, Vila Velha-ES, Brazil. Technical procedures, acquisition of data, manuscript preparation and writing
| | - Emy Hiura
- Veterinary Medical, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, UVV, Vila Velha-ES, Brazil. Technical procedures, acquisition of data, manuscript preparation and writing
| | - Wagner Alexey Back Fiorio
- Veterinay Medical, Advanced Imaging Center and Veterinary Pathology, Vila Velha-ES, Brazil. Histopathological examinations
| | - Gisele Pereira Valdetaro
- Veterinary Medical, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, UVV, Vila Velha-ES, Brazil. Technical procedures, acquisition of data, manuscript preparation and writing
| | - Dariele Vieira Gonçalves
- Veterinary Medical, Stem Cell and Cellular Therapy Laboratory, Animal Science Program, UVV, Vila Velha-ES, Brazil. Technical procedures, acquisition of data, manuscript preparation and writing
| | - Caroline Saraiva Silva
- Veterinay Medical, PetScan Diagnoses Center, Vila Velha-ES, Brazil. Imagens examinations
| | - Tatiana Champion
- Assistant Professor, Veterinary Department, Universidade Federal da Fronteira Sul (UFFS), Realeza-PR, Brazil. Statistics analysis, critical revision, final approval
| | - Daniela Campagnol
- Veterinay Medical, Animal Experimentation Division, Clinical Hospital, Universidade Federal do Rio Grande do Sul (UFRS), Porto Alegre-RS, Brazil. Substantive scientific and intellectual contributions to the study, critical revision
| |
Collapse
|
45
|
Zhang S, Liu Y, Derakhshanfar S, He W, Huang Q, Dong S, Rao J, Luo G, Zhong W, Liao W, Shi M, Xing M. Polymer Self-Assembled BMSCs with Cancer Tropism and Programmed Homing. Adv Healthc Mater 2018; 7:e1800118. [PMID: 30345648 DOI: 10.1002/adhm.201800118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/09/2018] [Indexed: 11/05/2022]
Abstract
Targeted therapy can improve the accuracy of diagnosis and treatment in the field of cancer management. Cellular surface engineering can enhance cell functions via mounting functional molecules onto cellular membranes. A novel amphiphilic hyperbranched polymer (AHP) conjugated with oleic acid (OA) and tumor-targeted ligand folic acid (FA) is employed. The lipophilic chain can self-assemble and infuse with the cytomembrane of bone marrow mesenchymal stem cells (BMSCs) with the end of FA left on the outside for targeting. The polymer tailored BMSCs can enhance tumor tropism in gastric cancer. BMSCs are characterized by the low immunogenicity and tumor tropism, which makes them promising targeting carriers. Regarding the integrated advantages of these two vectors, it is demonstrated that the functional amphiphilic AHP-OA-FA enhances the tumor tropism of BMSCs. Flow cytometry, standard MTT assay, and wound-healing assay show that AHP-OA-FA has no influence on CD expression, proliferative capacity, and cell motility of BMSCs, respectively. Furthermore, in vitro transwell assay and ex vivo fluorescence image verify that AHP-OA-FA enhances tumor tropism of BMSCs compared to BMSCs and AHP-OA-Rhodamine B-BMSCs. Finally, histological analysis demonstrates that AHP-OA-FA causes no damage to major organs. The results of this study suggest that living BMSCs self-assembled with a polymer might be a promising vehicle for targeted delivery to cancer cells.
Collapse
Affiliation(s)
- Shuyi Zhang
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Yuqing Liu
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical University Guangzhou 510006 China
- Burns InstituteSouthwest Hospital Chongqing 400038 China
| | - Soroosh Derakhshanfar
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Wanming He
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Qiong Huang
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Shumin Dong
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Jinjun Rao
- Key Laboratory of New Drug Screening of Guangdong ProvinceSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou 510006 China
| | - Gao‐Xing Luo
- Burns InstituteSouthwest Hospital Chongqing 400038 China
| | - Wen Zhong
- Department of Biosystem EngineeringUniversity of Manitoba Winnipeg R3T 5V6 Canada
| | - Wangjun Liao
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Min Shi
- Department of OncologyNanfang HospitalSouthern Medical University Guangzhou 510006 China
| | - Malcolm Xing
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical University Guangzhou 510006 China
- Burns InstituteSouthwest Hospital Chongqing 400038 China
- Department of Mechanical EngineeringDepartment of Biochemistry and Medical GeneticsUniversity of Manitoba Winnipeg R3T 5V6 Canada
| |
Collapse
|
46
|
Abstract
Inflammatory bowel disease (IBD) has evoked significant interest in human immunobiology given its tactical immune evasion methodologies resulting in acute immune destabilization. IBD comprising Crohn’s disease and Ulcerative colitis manifests as chronic inflammation in the gut mucosa, leading to complexities involving immune dysregulation in the T helper lymphocyte arm, effecting disease pathogenicity. The mucosa of the alimentary canal is constantly exposed to a myriad of food antigens and luminal microorganisms for which a consistent host-protective mechanism is operative in healthy people. Lowered mucosal immune expression which allows penetration of the epithelial barrier by infective pathogenic microbes elicits both innate and adaptive immune responses in the gut, culminating in aberrant intestinal inflammation. Interestingly, the IBD leukocyte repertoire is significantly entwined with chemokine-assisted chemotactic navigation into the sites of inflammation, which is also thought to generate favorable immune-suppressive responses. The functions of the cognate chemokine receptor, CCR6, which binds with its unique ligand CCL20, are expected to tilt the balance between upregulation of homeostatic tolerance and inflammatory pathophysiology. This review aims to critically examine the CCR6-driven immune pathways: TH1/TH2, TH1/TH17, TH17/Treg, IL-23/IL-17, Akt/ERK-1/2, ILC3, and TH9/TH2 for systematic investigation of its underlying mechanisms in the future and to underpin its importance in resolving IBD pathology. Thus, CCR6 occupies an exclusive position in gut immunology which renders it an invaluable therapeutic tool for the production of novel medicaments to treat IBD.
Collapse
|
47
|
van Rhijn-Brouwer FCC, Gremmels H, Fledderus JO, Schuurman AH, Bonte-Mineur F, Vonk MC, Voskuyl AE, de Vries-Bouwstra JK, Coert JH, Radstake TRDJ, van Laar JM, Verhaar MC. A randomised placebo-controlled double-blind trial to assess the safety of intramuscular administration of allogeneic mesenchymal stromal cells for digital ulcers in systemic sclerosis: the MANUS Trial protocol. BMJ Open 2018; 8:e020479. [PMID: 30127049 PMCID: PMC6104757 DOI: 10.1136/bmjopen-2017-020479] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an autoimmune disease characterised by inflammation, fibrosis and vasculopathy. Digital ulcers (DUs) are a frequent manifestation of vasculopathy in patients with SSc. Despite recent advances in pharmacological treatments, DU still have major health and economic implications. As there is currently no proven therapeutic strategy to promote DU healing, new treatments are urgently needed. Mesenchymal stem or stromal cells (MSCs) may provide a novel therapy for DU in SSc, because of their immunomodulatory and vasculoregenerative properties. Allogeneic MSC therapy involves functionally competent MSCs from healthy donors and may be used as 'off-the-shelf' available treatment. This study will evaluate whether allogeneic MSC therapy is a safe and potentially efficacious treatment for DU of SSc. METHODS AND ANALYSIS The MANUS (Mesenchymal stromal cells for Angiogenesis and Neovascularization in digital Ulcers of Systemic Sclerosis) Trial is a double-blind randomised placebo-controlled trial. 20 patients with SSc with refractory DU will be randomised to receive eight intramuscular injections with either placebo or 50*106 MSCs. The primary outcome is the toxicity of the treatment at 12 weeks after administration. Secondary outcomes include (serious) adverse events, number and time to healing of DU, pain, reported hand function, quality of life and SSc disease activity. We will also evaluate changes in nailfold capillaroscopy pattern, as well as biochemical parameters and biomarkers in peripheral blood and skin biopsies. Follow-up visits will be scheduled at 48 hours and 2, 4, 8, 12, 24 and 52 weeks post-treatment. If the results confirm safety, feasibility and potential efficacy, a large multicentre randomised controlled trial with longer follow-up will be initiated focusing on efficacy. ETHICS AND DISSEMINATION The study has been approved by the Dutch Central Committee on Research Concerning Human Subjects (protocol no: NL51705.000.15). The results will be disseminated through patient associations and conventional scientific channels. TRIAL REGISTRATION NUMBER NCT03211793; Pre-results.
Collapse
Affiliation(s)
| | - Hendrik Gremmels
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joost O Fledderus
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arnold H Schuurman
- Department of Plastic Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Femke Bonte-Mineur
- Department of Rheumatology, Maasstad Ziekenhuis, Rotterdam, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexandre E Voskuyl
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | - J Henk Coert
- Department of Plastic Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacob M van Laar
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
48
|
Trans-differentiation Induction of Human-mesenchymal Stem Cells Derived from Different Tissue Origin and Evaluation of their Potential for Differentiation into Corneal Epithelial-like Cells. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2018. [DOI: 10.12750/jet.2018.33.2.85] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
49
|
Nooshabadi VT, Mardpour S, Yousefi-Ahmadipour A, Allahverdi A, Izadpanah M, Daneshimehr F, Ai J, Banafshe HR, Ebrahimi-Barough S. The extracellular vesicles-derived from mesenchymal stromal cells: A new therapeutic option in regenerative medicine. J Cell Biochem 2018; 119:8048-8073. [PMID: 29377241 DOI: 10.1002/jcb.26726] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult multipotent cells that due to their ability to homing to damaged tissues and differentiate into specialized cells, are remarkable cells in the field of regenerative medicine. It's suggested that the predominant mechanism of MSCs in tissue repair might be related to their paracrine activity. The utilization of MSCs for tissue repair is initially based on the differentiation ability of these cells; however now it has been revealed that only a small fraction of the transplanted MSCs actually fuse and survive in host tissues. Indeed, MSCs supply the microenvironment with the secretion of soluble trophic factors, survival signals and the release of extracellular vesicles (EVs) such as exosome. Also, the paracrine activity of EVs could mediate the cellular communication to induce cell-differentiation/self-renewal. Recent findings suggest that EVs released by MSCs may also be critical in the physiological function of these cells. This review provides an overview of MSC-derived extracellular vesicles as a hopeful opportunity to advance novel cell-free therapy strategies that might prevail over the obstacles and risks associated with the use of native or engineered stem cells. EVs are very stable; they can pass the biological barriers without rejection and can shuttle bioactive molecules from one cell to another, causing the exchange of genetic information and reprogramming of the recipient cells. Moreover, extracellular vesicles may provide therapeutic cargo for a wide range of diseases and cancer therapy.
Collapse
Affiliation(s)
| | - Soura Mardpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Izadpanah
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Daneshimehr
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid R Banafshe
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Soontararak S, Chow L, Johnson V, Coy J, Wheat W, Regan D, Dow S. Mesenchymal Stem Cells (MSC) Derived from Induced Pluripotent Stem Cells (iPSC) Equivalent to Adipose-Derived MSC in Promoting Intestinal Healing and Microbiome Normalization in Mouse Inflammatory Bowel Disease Model. Stem Cells Transl Med 2018; 7:456-467. [PMID: 29635868 PMCID: PMC5980202 DOI: 10.1002/sctm.17-0305] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular therapy with allogeneic or autologous mesenchymal stem cells (MSC) has emerged as a promising new therapeutic strategy for managing inflammatory bowel disease (IBD). However, MSC therapy ideally requires a convenient and relatively homogenous cell source (typically bone marrow or adipose tissues) and the ability to generate cells with stable phenotype and function. An alternative means of generating allogeneic MSC is to derive them from induced pluripotent stem cells (iPSC), which could in theory provide an indefinite supply of MSC with well-defined phenotype and function. Therefore, we compared the effectiveness of iPSC-derived MSC (iMSC) and adipose-derived MSC (adMSC) in a mouse model of IBD (dextran sodium sulfate-induced colitis), and investigated mechanisms of intestinal protection. We found that iMSC were equivalent to adMSC in terms of significantly improving clinical abnormalities in treated mice and reducing lesion scores and inflammation in the gut. Administration of iMSC also stimulated significant intestinal epithelial cell proliferation, increased in the numbers of Lgr5+ intestinal stem cells, and increased intestinal angiogenesis. In addition, the microbiome alterations present in mice with colitis were partially restored to resemble those of healthy mice following treatment with iMSC or adMSC. Thus, iMSC administration improved overall intestinal health and healing with equivalent potency to treatment with adMSC. This therefore is the first report of the effectiveness of iMSC in the treatment of IBD, along with a description of unique mechanisms of action with respect to intestinal healing and microbiome restoration. Stem Cells Translational Medicine 2018;7:456-467.
Collapse
Affiliation(s)
- Sirikul Soontararak
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Lyndah Chow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Jonathan Coy
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - William Wheat
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Daniel Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|