151
|
Cytokines Differently Define the Immunomodulation of Mesenchymal Stem Cells from the Periodontal Ligament. Cells 2020; 9:cells9051222. [PMID: 32423044 PMCID: PMC7290931 DOI: 10.3390/cells9051222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) play an important role in periodontal tissue homeostasis and regeneration. The function of these cells in vivo depends largely on their immunomodulatory ability, which is reciprocally regulated by immune cells via cytokines, particularly interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β. Different cytokines activate distinct signaling pathways and might differently affect immunomodulatory activities of hPDLSCs. This study directly compared the effect of IFN-γ, TNF-α, or IL-1β treated primary hPDLSCs on allogenic CD4+ T lymphocyte proliferation and apoptosis in an indirect co-culture model. The effects of IFN-γ, TNF-α, and IL-1β on the expression of specific immunomodulatory factors such as intoleamine-2,3-dioxygenase-1 (IDO-1), prostaglandin E2 (PGE2), and programmed cell death 1 ligand 1 (PD-L1) and ligand 2 (PD-L2) in hPDLSCs were compared. The contribution of different immunomodulatory mediators to the immunomodulatory effects of hPDLSCs in the indirect co-culture experiments was assessed using specific inhibitors. Proliferation of CD4+ T lymphocytes was inhibited by hPDLSCs, and this effect was strongly enhanced by IFN-γ and IL-1β but not by TNF-α. Apoptosis of CD4+ T lymphocytes was decreased by hPDLSCs per se. This effect was counteracted by IFN-γ or IL-1β. Additionally, IFN-γ, TNF-α, and IL-1β differently regulated all investigated immunomediators in hPDLSCs. Pharmacological inhibition of immunomediators showed that their contribution in regulating CD4+ T lymphocytes depends on the cytokine milieu. Our data indicate that inflammatory cytokines activate specific immunomodulatory mechanisms in hPDLSCs and the expression of particular immunomodulatory factors, which underlies a complex reciprocal interaction between hPDLSCs and CD4+ T lymphocytes.
Collapse
|
152
|
Li X, Li J, Lu F, Cao Y, Xing J, Li J, Hou R, Yin G, Zhang K. Role of SPRED1 in keratinocyte proliferation in psoriasis. J Dermatol 2020; 47:735-742. [PMID: 32396270 DOI: 10.1111/1346-8138.15369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/05/2020] [Indexed: 12/13/2022]
Abstract
Psoriasis is a recurrent inflammatory skin disease, affecting approximately 2% of the population. Previous studies have demonstrated that psoriatic dermal mesenchymal stem cells (DMSC) stimulated keratinocyte (KC) proliferation and that psoriasis exhibited missense SPRED1 mutations. To further investigate the molecular mechanism by which psoriatic DMSC stimulate KC proliferation, and the role of missense SPRED1 mutations in psoriasis, we assessed expression levels of miRNA, and both mRNA and protein of SPRED1 in normal human epidermal keratinocyte cells (NHEK) cocultured with either psoriatic or control DMSC. Expression levels of miRNA and mRNA were determined by RNA sequencing. Expression levels of spred1 protein were assessed using western blot analysis. Moreover, the variation in SPRED1 was also examined by whole-genome sequencing in 665 psoriatic patients, and verified by Sanger sequencing. Our results showed that coculture of NHEK with psoriatic DMSC induced 32 differentially expressed miRNA, in which expression levels of miR-1 increased approximately 16-fold over control DMSC-treated NHEK (P < 0.05). Likewise, expression levels of miR-21-3p increased over twofold (P < 0.05). Moreover, coculture of NHEK with psoriatic DMSC induced marked increase in expression levels of mRNA for MAPK3, CDC25B and CDC25C, while decreasing expression levels of SPRED1 mRNA and protein in comparison with control DMSC treatment (P < 0.05 for all between cocultured with control and psoriatic DMSC). Furthermore, psoriasis displayed non-synonymous mutation of SPRED1 enriched in exon 7: c.A881T:p.Y294F (chr15:38351210). These results suggest that dysregulation and mutations of SPRED1 may participate in the pathogenesis of psoriasis, including epidermal hyperproliferation.
Collapse
Affiliation(s)
- Xinhua Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Funa Lu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Yue Cao
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianxiao Xing
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Juan Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruixia Hou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Guohua Yin
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
153
|
Identifying the Therapeutic Significance of Mesenchymal Stem Cells. Cells 2020; 9:cells9051145. [PMID: 32384763 PMCID: PMC7291143 DOI: 10.3390/cells9051145] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
The pleiotropic behavior of mesenchymal stem cells (MSCs) has gained global attention due to their immense potential for immunosuppression and their therapeutic role in immune disorders. MSCs migrate towards inflamed microenvironments, produce anti-inflammatory cytokines and conceal themselves from the innate immune system. These signatures are the reason for the uprising in the sciences of cellular therapy in the last decades. Irrespective of their therapeutic role in immune disorders, some factors limit beneficial effects such as inconsistency of cell characteristics, erratic protocols, deviating dosages, and diverse transfusion patterns. Conclusive protocols for cell culture, differentiation, expansion, and cryopreservation of MSCs are of the utmost importance for a better understanding of MSCs in therapeutic applications. In this review, we address the immunomodulatory properties and immunosuppressive actions of MSCs. Also, we sum up the results of the enhancement, utilization, and therapeutic responses of MSCs in treating inflammatory diseases, metabolic disorders and diabetes.
Collapse
|
154
|
Mendoza-Reinoso V, McCauley LK, Fournier PG. Contribution of Macrophages and T Cells in Skeletal Metastasis. Cancers (Basel) 2020; 12:E1014. [PMID: 32326073 PMCID: PMC7226332 DOI: 10.3390/cancers12041014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Bone is a common site for metastases with a local microenvironment that is highly conducive for tumor establishment and growth. The bone marrow is replete with myeloid and lymphoid linage cells that provide a fertile niche for metastatic cancer cells promoting their survival and growth. Here, we discuss the role of macrophages and T cells in pro- and anti-tumoral mechanisms, their interaction to support cancer cell growth, and their contribution to the development of skeletal metastases. Importantly, immunotherapeutic strategies targeting macrophages and T cells in cancer are also discussed in this review as they represent a great promise for patients suffering from incurable bone metastases.
Collapse
Affiliation(s)
- Veronica Mendoza-Reinoso
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (V.M.-R.); (L.K.M.)
| | - Laurie K. McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (V.M.-R.); (L.K.M.)
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pierrick G.J. Fournier
- Biomedical Innovation Department, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada, BC 22860, Mexico
| |
Collapse
|
155
|
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol 2020; 11:639. [PMID: 32328072 PMCID: PMC7160320 DOI: 10.3389/fimmu.2020.00639] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular Disease (CVD) is a leading cause of mortality within the United States. Current treatments being administered to patients who suffered a myocardial infarction (MI) have increased patient survival, but do not facilitate the replacement of damaged myocardium. Recent studies demonstrate that stem cell-based therapies promote myocardial repair; however, the poor engraftment of the transferred stem cell populations within the infarcted myocardium is a major limitation, regardless of the cell type. One explanation for poor cell retention is attributed to the harsh inflammatory response mounted following MI. The inflammatory response coupled to cardiac repair processes is divided into two distinct phases. The first phase is initiated during ischemic injury when necrosed myocardium releases Danger Associated Molecular Patterns (DAMPs) and chemokines/cytokines to induce the activation and recruitment of neutrophils and pro-inflammatory M1 macrophages (MΦs); in turn, facilitating necrotic tissue clearance. During the second phase, a shift from the M1 inflammatory functional phenotype to the M2 anti-inflammatory and pro-reparative functional phenotype, permits the resolution of inflammation and the establishment of tissue repair. T-regulatory cells (Tregs) are also influential in mediating the establishment of the pro-reparative phase by directly regulating M1 to M2 MΦ differentiation. Current studies suggest CD4+ T-lymphocyte populations become activated when presented with autoantigens released from the injured myocardium. The identity of the cardiac autoantigens or paracrine signaling molecules released from the ischemic tissue that directly mediate the phenotypic plasticity of T-lymphocyte populations in the post-MI heart are just beginning to be elucidated. Stem cells are enriched centers that contain a diverse paracrine secretome that can directly regulate responses within neighboring cell populations. Previous studies identify that stem cell mediated paracrine signaling can influence the phenotype and function of immune cell populations in vitro, but how stem cells directly mediate the inflammatory microenvironment of the ischemic heart is poorly characterized and is a topic of extensive investigation. In this review, we summarize the complex literature that details the inflammatory microenvironment of the ischemic heart and provide novel insights regarding how paracrine mediated signaling produced by stem cell-based therapies can regulate immune cell subsets to facilitate pro-reparative myocardial wound healing.
Collapse
Affiliation(s)
- Marcus J Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
156
|
Li X, Xu J, Li P. Rat bone mesenchymal stem cells exert antiproliferative effects on nicotine‑exposed T cells via iNOS production. Mol Med Rep 2020; 21:2267-2275. [PMID: 32186760 DOI: 10.3892/mmr.2020.11027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/14/2020] [Indexed: 11/05/2022] Open
Abstract
Adoptive transfer of bone marrow‑derived mesenchymal stem cells (BMSCs) significantly alleviates smoking‑induced chronic obstructive pulmonary disease (COPD) in rats. Considering the critical roles of T cells during COPD development, the present study aimed to further identify the molecular mechanisms underlying the antiproliferative effect of BMSCs on splenic T cells isolated from rats following chronic exposure to nicotine. Splenic T cells were co‑cultured with rat BMSCs at various ratios and subsequently, T‑cell proliferation was measured using the Cell Counting Kit‑8 assay. The effects of the inducible nitric oxide synthase (iNOS) inhibitor N‑nitro‑L‑arginine methylester (L‑NAME) and short hairpin (sh)RNA‑lentivirus‑mediated knockdown of iNOS in BMSCs on T‑cell proliferation were evaluated. The expression levels of iNOS and STAT5 phosphorylation in BMSCs and T cells, respectively, were assessed by reverse transcription‑quantitative PCR and western blotting. A higher ratio of BMSCs to T cells resulted in increased inhibition of T‑cell proliferation; therefore, the ratio of 1:20 was selected for further in vitro experiments. At a dose of 5 µM, L‑NAME displayed the strongest ability to reverse the antiproliferative effects of BMSCs in the co‑culture system. Both L‑NAME treatment and shRNA‑mediated knockdown of iNOS expression significantly decreased the suppressive effects of BMSCs, downregulated iNOS expression at the mRNA and protein levels in BMSCs, and enhanced STAT5 phosphorylation in T cells. BMSCs inhibited the proliferation of nicotine‑exposed T cells, which was associated with iNOS expression in BMSCs and decreased STAT5 phosphorylation in T cells. The present study indicated the potential mechanisms underlying the beneficial effects of BMSC infusion in patients with chronic smoking‑induced COPD and emphysema.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Respiratory and Critical Care Medicine, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Jianying Xu
- Department of Respiratory and Critical Care Medicine, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Pingping Li
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi 030009, P.R. China
| |
Collapse
|
157
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
158
|
Zhao H, Xie L, Clemens JL, Zong L, McLane MW, Arif H, Feller MC, Jia B, Zhu Y, Facciabene A, Ozen M, Lei J, Burd I. Mouse Bone Marrow-Derived Mesenchymal Stem Cells Alleviate Perinatal Brain Injury Via a CD8 + T Cell Mechanism in a Model of Intrauterine Inflammation. Reprod Sci 2020; 27:1465-1476. [PMID: 31997258 DOI: 10.1007/s43032-020-00157-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
The objective of this study was to determine if mouse bone marrow-derived mesenchymal stem cells (BMMSCs) ameliorate preterm birth and perinatal brain injury induced by intrauterine inflammation (IUI). A mouse model of IUI-induced perinatal brain injury at embryonic (E) day 17 was utilized. BMMSCs were derived from GFP-transgenic mice and phenotypically confirmed to be CD44+, Sca-1+, CD45-, CD34-, CD11b-, and CD11c- by flow cytometry and sorted by fluorescence-activated cell sorting (FACS). Dams were assigned to four groups: phosphate-buffered saline (PBS) + PBS, PBS + BMMSCs, lipopolysaccharide (LPS) + PBS, and LPS + BMMSCs. Following maternal IUI, there was a significant increase in CD8+ T cells in the placentas. Maternally administered BMMSCs trafficked to the fetal side of the placenta and resulted in significantly decreased placental CD8+ T cells. Furthermore, fetal trafficking of maternally administered BMMSCs correlated with an improved performance on offspring neurobehavioral testing in LPS + BMMSC group compared with LPS + PBS group. Our data support that maternal administration of BMMSCs can alleviate perinatal inflammation-induced brain injury and improve neurobehavioral outcomes in the offspring via CD8+ T cell immunomodulation at the feto-placental interface.
Collapse
Affiliation(s)
- Hongxi Zhao
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Li Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Julia L Clemens
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Lu Zong
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Michael W McLane
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Hattan Arif
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Mia C Feller
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Bei Jia
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yan Zhu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Andreas Facciabene
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Maide Ozen
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
159
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
160
|
Rostami Z, Khorashadizadeh M, Naseri M. Immunoregulatory properties of mesenchymal stem cells: Micro-RNAs. Immunol Lett 2020; 219:34-45. [PMID: 31917251 DOI: 10.1016/j.imlet.2019.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that are excellent candidates for different cellular therapies due to their physiological properties such as immunoregulatory function. whetheare currently utilized for regenerative medication and treatment of a number of inflammatory illnesses given their ability to considerably impact tissue microenvironments via extracellular vesicles or toll-like receptor pathway modulation. MicroRNAs (miRNAs) are small noncoding RNAs that target the messenger RNA and play a critical role in different biological procedures, such as the development and reaction of the immune system. Moreover, miRNAs have recently been revealed to have serious functions in MSCs to regulate immunomodulatory properties. In this review, we study how the miRNAs pathway can modulate the immunoregulatory activity of MSCs by counting their interactions with immune cells and also discuss the possibility of using miRNA-based implications for MSC-based therapies.
Collapse
Affiliation(s)
- Zeinab Rostami
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Medical Biotechnology (PhD), Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran; Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
161
|
Feng Y, Wang AT, Jia HH, Zhao M, Yu H. A Brief Analysis of Mesenchymal Stem Cells as Biological Drugs for the Treatment of Acute-on-Chronic Liver Failure (ACLF): Safety and Potency. Curr Stem Cell Res Ther 2020; 15:202-210. [PMID: 31893994 DOI: 10.2174/1574888x15666200101124317] [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] [Received: 10/06/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Abstract
Acute-on-Chronic Liver Failure (ACLF) is characterized by acute exacerbation of chronic hepatitis, organ failure, high mortality, and poor prognosis. At present, the clinical methods of treatment include comprehensive treatment with medicines, artificial liver system, and Orthotopic Liver Transplantation (OLT), and of these, OLT is considered the most effective treatment for ACLF. However, it is difficult for ACLF patients to benefit from OLT due to the shortage of liver donors, high cost, unpredictable postoperative complications, and long-term use of immunosuppressive drugs; therefore, it is important to explore a new treatment option. With the development of stem cell transplantation technology in recent years, several studies have shown that treatment of ACLF with Mesenchymal Stem Cells (MSCs) leads to higher survival rates, and has good tolerance and safety rates, thereby improving the liver function and quality of life of patients; it has also become one of the popular research topics in clinical trials. This paper summarizes the current clinical interventions and treatments of ACLF, including the clinical trials, therapeutic mechanisms, and research progress on MSC application in the treatment of ACLF. The problems and challenges of the development of MSC-based therapy in the future are also discussed.
Collapse
Affiliation(s)
- Ying Feng
- Cell Products of National Engineering Research Center, Tianjin 300457, China.,National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Ai-Tong Wang
- Cell Products of National Engineering Research Center, Tianjin 300457, China.,National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hong-Hong Jia
- Cell Products of National Engineering Research Center, Tianjin 300457, China.,National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Meng Zhao
- Cell Products of National Engineering Research Center, Tianjin 300457, China.,National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hao Yu
- Cell Products of National Engineering Research Center, Tianjin 300457, China.,National Stem Cell Engineering Research Center, Tianjin 300457, China
| |
Collapse
|
162
|
Hejretová L, Čedíková M, Dolejšová M, Vlas T, Jindra P, Lysák D, Holubová M. Comparison of the immunomodulatory effect of single MSC batches versus pooled MSC products. Cell Tissue Bank 2019; 21:119-129. [PMID: 31863261 DOI: 10.1007/s10561-019-09805-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Severe corticosteroid-refractory graft-versus-host-disease (GVHD) is a major non-relapse cause of mortality and morbidity after an allogeneic hematopoietic stem cell transplantation (allo-HSCT). One of the most promising treatment options is using advanced therapy medicinal products based on mesenchymal stem cells (MSCs) immunomodulation ability. The protocols of MSC application differ in many parameters including a source of MSC, a dose, a number of doses or way of preparation of the medicinal product. The process is limited by the need for laborious and expensive manufacturing processes fraught with batch-to-batch variability. In our study, we compared the immunomodulatory effects of different MSC batches versus pooled MSC, specifically the influence on lymphocyte proliferation, the metabolic activity, and the expression of activation markers on T cells. Our goal was to determine whether the effect depends on donor-to-donor heterogeneity and if pooling of MSCs could increase their immunomodulatory ability. All tested batches showed an immunomodulatory effect, with no significant differences between the groups. Our study suggests that immunosuppressive potential is comparable in single batches and pooled products, and the use of products got from individual donors is suitable to treat corticosteroid-refractory GVHD.
Collapse
Affiliation(s)
- L Hejretová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Čedíková
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - M Dolejšová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - T Vlas
- Institute of Immunology and Allergology, University Hospital and Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - P Jindra
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - D Lysák
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - M Holubová
- Department of Haematology and Oncology, University Hospital, Pilsen, Alej Svobody 80, 304 60, Pilsen, Czech Republic. .,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| |
Collapse
|
163
|
Anti-Inflammatory Action of Heterogeneous Nuclear Ribonucleoprotein A2/B1 in Patients with Autoimmune Endocrine Disorders. J Clin Med 2019; 9:jcm9010009. [PMID: 31861546 PMCID: PMC7019344 DOI: 10.3390/jcm9010009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/27/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022] Open
Abstract
Our previous studies documented that human fibroblast-limbal stem cells (f-LSCs) possess immunosuppressive capabilities, playing a role in regulating T-cell activity. This study highlights the molecular activities by which human f-LSCs can attenuate the inflammatory responses of self-reactive peripheral blood mononuclear cells (PBMCs) collected from patients with autoimmune endocrine diseases (AEDs). Anti-CD3 activated PBMCs from twenty healthy donors and fifty-two patients with AEDs were cocultured on f-LSC monolayer. 2D-DIGE proteomic experiments, mass spectrometry sequencing and functional in vitro assays were assessed in cocultured PBMCs. We identified the downmodulation of several human heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) isoforms in healthy and AED activated PBMCs upon f-LSC interaction. The reduction of hnRNPA2/B1 protein expression largely affected the cycling ki67+, CD25+, PD-1+ reactive cells and the double marked CD8+/hnRNPA2B1+ T cell subset. Anti-PD1 blocking experiments evoked hnRNPA2/B1 overexpression, attributing putative activation function to the protein. hnRNPA2/B2 transient silencing inverted immunopolarization of the self-reactive PBMCs from AEDs toward a M2/Th2-type background. Pharmacological inhibition and co-immunoprecipitation experiments demonstrated the involvement of NF-ĸB in hnRNPA2/B activity and turnover. Our data indicate cardinal involvement of hnRNP A2/B1 protein in peripheral mechanisms of tolerance restoration and attenuation of inflammation, identifying a novel immunoplayer potentially targetable in all AEDs.
Collapse
|
164
|
Hu C, Li L. The immunoregulation of mesenchymal stem cells plays a critical role in improving the prognosis of liver transplantation. J Transl Med 2019; 17:412. [PMID: 31823784 PMCID: PMC6905033 DOI: 10.1186/s12967-019-02167-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
The liver is supplied by a dual blood supply, including the portal venous system and the hepatic arterial system; thus, the liver organ is exposed to multiple gut microbial products, metabolic products, and toxins; is sensitive to extraneous pathogens; and can develop liver failure, liver cirrhosis and hepatocellular carcinoma (HCC) after short-term or long-term injury. Although liver transplantation (LT) serves as the only effective treatment for patients with end-stage liver diseases, it is not very popular because of the complications and low survival rates. Although the liver is generally termed an immune and tolerogenic organ with adaptive systems consisting of humoral immunity and cell-mediated immunity, a high rejection rate is still the main complication in patients with LT. Growing evidence has shown that mesenchymal stromal cell (MSC) transplantation could serve as an effective immunomodulatory strategy to induce tolerance in various immune-related disorders. MSCs are reported to inhibit the immune response from innate immune cells, including macrophages, dendritic cells (DCs), natural killer cells (NK cells), and natural killer T (NKT) cells, and that from adaptive immune cells, including T cells, B cells and other liver-specific immune cells, for the generation of a tolerogenic microenvironment. In this review, we summarized the relationship between LT and immunoregulation, and we focused on how to improve the effects of MSC transplantation to improve the prognosis of LT. Only after exhaustive clarification of the potential immunoregulatory mechanisms of MSCs in vitro and in vivo can we implement MSC protocols in routine clinical practice to improve LT outcome.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
165
|
Okamura A, Matsushita T, Komuro A, Kobayashi T, Maeda S, Hamaguchi Y, Takehara K. Adipose-derived stromal/stem cells successfully attenuate the fibrosis of scleroderma mouse models. Int J Rheum Dis 2019; 23:216-225. [PMID: 31808305 DOI: 10.1111/1756-185x.13764] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/10/2019] [Accepted: 11/15/2019] [Indexed: 12/13/2022]
Abstract
AIM Systemic sclerosis (SSc) is an autoimmune disease characterized by skin and lung fibrosis. Although SSc has a high mortality risk, an effective treatment for the disease has not been established yet. Mesenchymal stromal/stem cells (MSCs) are multipotential nonhematopoietic progenitor cells that have the ability to regulate immune responses. Adipose-derived stromal/stem cells (ASCs), one of the types of MSCs, have the advantage of accessibility and potent immunomodulatory effects when compared with other MSCs, such as bone marrow-derived MSCs. This study aimed to investigate the antifibrotic effect of ASCs in scleroderma mouse models, including bleomycin-induced scleroderma and sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) models. METHOD ASCs were intravenously administered to a bleomycin-induced scleroderma or Scl-cGVHD model on day 0. We compared the skin and lung fibrosis of scleroderma model mice between the ASC-treated group and control group. RESULTS Administration of ASCs attenuated the skin and lung fibrosis of bleomycin-induced scleroderma and Scl-cGVHD model mice compared to that in the control mice. Immunohistochemical staining showed that ASCs suppressed the infiltration of CD4+ , CD8+ T cells and macrophages into the dermis of bleomycin model mice compared to that in control mice. In addition, ASCs attenuated the messenger RNA expression of collagen and fibrogenic cytokines, such as interleukin (IL)-6 and IL-13, in the skin of bleomycin model mice. ASCs also reduced the frequency of fibrogenic cytokine-producing CD4+ T cells and effector B cells in the spleen of bleomycin model mice. CONCLUSION ASCs could prove to be a potential therapeutic agent for use in patients with SSc.
Collapse
Affiliation(s)
- Ai Okamura
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Plastic Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Takashi Matsushita
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Akito Komuro
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Plastic Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Tadahiro Kobayashi
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shintaro Maeda
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yasuhito Hamaguchi
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kazuhiko Takehara
- Department of Dermatology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
166
|
Godoy JAP, Paiva RMA, Souza AM, Kondo AT, Kutner JM, Okamoto OK. Clinical Translation of Mesenchymal Stromal Cell Therapy for Graft Versus Host Disease. Front Cell Dev Biol 2019; 7:255. [PMID: 31824942 PMCID: PMC6881464 DOI: 10.3389/fcell.2019.00255] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
Graft versus host disease (GVHD) is a common condition in patients subjected to allogeneic hematopoietic stem cell transplantation (HSCT). The immune cells derived from the grafted stem cells attack recipient's tissues, including those from the skin, liver, eyes, mouth, lungs, gastrointestinal tract, neuromuscular system, and genitourinary tract, may lead to severe morbidity and mortality. Acute GVHD can occur within few weeks after the allogeneic cells have engrafted in the recipient while chronic GVHD may occur any time after transplant, typically within months. Although treatable by systemic corticosteroid administration, effective responses are not achieved for a significant proportion of patients, a condition associated with poor prognosis. The use of multipotent mesenchymal stromal cells (MSCs) as an alternative to treat steroid-refractory GVHD had improved last decade, but the results are still controversial. Some studies have shown improvement in the life quality of patients after MSCs treatment, while others have found no significant benefits. In addition to variations in trial design, discrepancies in protocols for MSCs isolation, characterization, and ex vivo manipulation, account for inconsistent clinical results. In this review, we discuss the immunomodulatory properties supporting the therapeutic use of MSCs in GVHD and contextualize the main clinical findings of recent trials using these cells. Critical parameters for the clinical translation of MSCs, including consistent production of MSCs according to Good Manufacturing Practices (GMPs) and informative potency assays for product quality control (QC), are addressed.
Collapse
Affiliation(s)
- Juliana A. P. Godoy
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Raquel M. A. Paiva
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Aline M. Souza
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andrea T. Kondo
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jose M. Kutner
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Oswaldo K. Okamoto
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
167
|
Castro LL, Kitoko JZ, Xisto DG, Olsen PC, Guedes HLM, Morales MM, Lopes-Pacheco M, Cruz FF, Rocco PRM. Multiple doses of adipose tissue-derived mesenchymal stromal cells induce immunosuppression in experimental asthma. Stem Cells Transl Med 2019; 9:250-260. [PMID: 31746562 PMCID: PMC6988761 DOI: 10.1002/sctm.19-0120] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022] Open
Abstract
In experimental house dust mite (HDM)‐induced allergic asthma, therapeutic administration of a single dose of adipose tissue‐derived mesenchymal stromal cells (MSCs) ameliorates lung inflammation but is unable to reverse remodeling. We hypothesized that multiple doses of MSCs might exert better therapeutic effects by reducing lung inflammation and remodeling but might also result in immunosuppressive effects in experimental asthma. HDM was administered intranasally in C57BL/6 mice. After the last HDM challenge, mice received two or three doses of MSCs (105 cells per day) or saline intravenously. An additional cohort of mice received dexamethasone as a positive control for immunosuppression. Two and three doses of MSCs reduced lung inflammation, levels of interleukin (IL)‐4, IL‐13, and eotaxin; total leukocyte, CD4+ T‐cell, and eosinophil counts in bronchoalveolar lavage fluid; and total leukocyte counts in bone marrow, spleen, and mediastinal lymph nodes. Two and three doses of MSCs also reduced collagen fiber content and transforming growth factor‐β levels in lung tissue; however, the three‐dose regimen was more effective, and reduced these parameters to control levels, while also decreasing α‐actin content in lung tissue. Two and three doses of MSCs improved lung mechanics. Dexamethasone, two and three doses of MSCs similarly increased galectin levels, but only the three‐dose regimen increased CD39 levels in the thymus. Dexamethasone and the three‐dose, but not the two‐dose regimen, also increased levels of programmed death receptor‐1 and IL‐10, while reducing CD4+CD8low cell percentage in the thymus. In conclusion, multiple doses of MSCs reduced lung inflammation and remodeling while causing immunosuppression in HDM‐induced allergic asthma.
Collapse
Affiliation(s)
- Ligia L Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Jamil Z Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Herbert L M Guedes
- Laboratory of Glycobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
168
|
Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif 2019; 53:e12712. [PMID: 31730279 PMCID: PMC6985662 DOI: 10.1111/cpr.12712] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/11/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be derived from various adult tissues with multipotent and self‐renewal abilities. The characteristics of presenting no major ethical concerns, having low immunogenicity and possessing immune modulation functions make MSCs promising candidates for stem cell therapies. MSCs could promote inflammation when the immune system is underactivated and restrain inflammation when the immune system is overactivated to avoid self‐overattack. These cells express many immune suppressors to switch them from a pro‐inflammatory phenotype to an anti‐inflammatory phenotype, resulting in immune effector cell suppression and immune suppressor cell activation. We would discuss the mechanisms governing the immune modulation function of these cells in this review, especially the immune‐suppressive effects of MSCs.
Collapse
Affiliation(s)
- Wei Jiang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
169
|
Medhat D, Rodríguez CI, Infante A. Immunomodulatory Effects of MSCs in Bone Healing. Int J Mol Sci 2019; 20:ijms20215467. [PMID: 31684035 PMCID: PMC6862454 DOI: 10.3390/ijms20215467] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into multilineage cells, thus making them a significant prospect as a cell source for regenerative therapy; however, the differentiation capacity of MSCs into osteoblasts seems to not be the main mechanism responsible for the benefits associated with human mesenchymal stem cells hMSCs when used in cell therapy approaches. The process of bone fracture restoration starts with an instant inflammatory reaction, as the innate immune system responds with cytokines that enhance and activate many cell types, including MSCs, at the site of the injury. In this review, we address the influence of MSCs on the immune system in fracture repair and osteogenesis. This paradigm offers a means of distinguishing target bone diseases to be treated with MSC therapy to enhance bone repair by targeting the crosstalk between MSCs and the immune system.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
170
|
Ciciarello M, Corradi G, Loscocco F, Visani G, Monaco F, Cavo M, Curti A, Isidori A. The Yin and Yang of the Bone Marrow Microenvironment: Pros and Cons of Mesenchymal Stromal Cells in Acute Myeloid Leukemia. Front Oncol 2019; 9:1135. [PMID: 31709192 PMCID: PMC6823864 DOI: 10.3389/fonc.2019.01135] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have, for a long time, been recognized as pivotal contributors in the set up and maintenance of the hematopoietic stem cell (HSC) niche, as well as in the development and differentiation of the lympho-hematopoietic system. MSCs also have a unique immunomodulatory capacity, which makes them able to affect, both in vitro and in vivo, the function of immune cells. These features, namely the facilitation of stem cell engraftment and the inhibition of lymphocyte responses, have both proven essential for successful allogeneic stem cell transplantation (allo-SCT), which remains the only curative option for several hematologic malignancies. For example, in steroid-refractory acute graft-vs. host disease developing after allo-SCT, MSCs have produced significant results and are now considered a treatment option. However, more recently, the other side of the MSC coin has been unveiled, because of their emerging role in creating a protective and immune-tolerant microenvironment able to support the survival of leukemic cells and affect the response to therapies. In this light, it has been proposed that the failure of current treatments to efficiently override the stroma-mediated protection of leukemic cells accounts for the high rate of relapse in acute myeloid leukemia, at least in part. In this review, we will focus on emerging microenvironment-driven mechanisms conferring a survival advantage to leukemic cells overt physiological HSCs. This body of evidence increasingly highlights the opportunity to consider tumor-microenvironment interactions when designing new therapeutic strategies.
Collapse
Affiliation(s)
- Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giulia Corradi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Federica Monaco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Antonio Curti
- Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| |
Collapse
|
171
|
Rahmatizadeh F, Gholizadeh-Ghaleh Aziz S, Khodadadi K, Lale Ataei M, Ebrahimie E, Soleimani Rad J, Pashaiasl M. Bidirectional and Opposite Effects of Naïve Mesenchymal Stem Cells on Tumor Growth and Progression. Adv Pharm Bull 2019; 9:539-558. [PMID: 31857958 PMCID: PMC6912184 DOI: 10.15171/apb.2019.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer has long been considered as a heterogeneous population of uncontrolled proliferation of
different transformed cell types. The recent findings concerning tumorigeneses have highlighted
the fact that tumors can progress through tight relationships among tumor cells, cellular, and
non-cellular components which are present within tumor tissues. In recent years, studies have
shown that mesenchymal stem cells (MSCs) are essential components of non-tumor cells within
the tumor tissues that can strongly affect tumor development. Several forms of MSCs have been
identified within tumor stroma. Naïve (innate) mesenchymal stem cells (N-MSCs) derived from
different sources are mostly recruited into the tumor stroma. N-MSCs exert dual and divergent
effects on tumor growth through different conditions and factors such as toll-like receptor
priming (TLR-priming), which is the primary underlying causes of opposite effects. Moreover,
MSCs also have the contrary effects by various molecular mechanisms relying on direct cellto-
cell connections and indirect communications through the autocrine, paracrine routes, and
tumor microenvironment (TME).
Overall, cell-based therapies will hold great promise to provide novel anticancer treatments.
However, the application of intact MSCs in cancer treatment can theoretically cause adverse
clinical outcomes. It is essential that to extensively analysis the effective factors and conditions
in which underlying mechanisms are adopted by MSCs when encounter with cancer.
The aim is to review the cellular and molecular mechanisms underlying the dual effects of
MSCs followed by the importance of polarization of MSCs through priming of TLRs.
Collapse
Affiliation(s)
- Faramarz Rahmatizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Khodadad Khodadadi
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, Melbourne, Australia
| | - Maryam Lale Ataei
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Ebrahimie
- Adelaide Medical School, University of Adelaide, Adelaide, Australia.,School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, Australia
| | - Jafar Soleimani Rad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Maryam Pashaiasl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran.,Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
172
|
Kong D, Liu X, Li X, Hu J, Li X, Xiao J, Dai Y, He M, Liu X, Jiang Y, Cui R, Zhang L, Wang J, Li A, Wang F, Zhang Y, Xiao J, Wang W, Zheng C. Mesenchymal stem cells significantly improved treatment effects of Linezolid on severe pneumonia in a rabbit model. Biosci Rep 2019; 39:BSR20182455. [PMID: 31484796 PMCID: PMC6746999 DOI: 10.1042/bsr20182455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate whether co-administration of mesenchymal stromal cells (MSC) and linezolid (LZD) into a rabbit model of methicillin-resistant Staphylococcus aureus (MRSA)-infected pneumonia would bring a synergistic therapeutic effect. Human umbilical cord-derived MSCs (hUMSCs) were isolated and characterized. A rabbit model of pneumonia was constructed by delivering 1 × 1010 CFU MRSA via a bronchoscope into the basal segment of lower lobe of right lung. Through analyzing vital sign, pulmonary auscultation, SpO2, chest imaging, bronchoscopic manifestations, pathology, neutrophil percentage, and inflammatory factors, we verified that a rabbit model of MRSA-induced pneumonia was successfully constructed. Individual treatment with LZD (50 mg/kg for two times/day) resulted in improvement of body weight, chest imaging, bronchoscopic manifestations, histological parameters, and IL-10 concentration in plasma (P<0.01), decreasing pulmonary auscultation, and reduction of IL-8, IL-6, CRP, and TNF-α concentrations in plasma (P<0.01) compared with the pneumonia model group at 48 and 168 h. Compared with LZD group, co-administration of hUMSCs (1 × 106/kg for two times at 6 and 72 h after MRSA instillation) and LZD further increased the body weight (P<0.05). The changes we observed from chest imaging, bronchoscopic manifestations and pathology revealed that co-administration of hUMSCs and LZD reduced lung inflammation more significantly than that of LZD group. The plasma levels of IL-8, IL-6, CRP, and TNF-α in combined group decreased dramatically compared with the LZD group (P<0.05). In conclusion, hUMSCs administration significantly improved therapeutic effects of LZD on pneumonia resulted from MRSA infection in a rabbit model.
Collapse
Affiliation(s)
- Dexiao Kong
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| | - Xia Liu
- Department of Respiratory Intervention, Qilu Children's Hospital of Shandong University, Jinan, Shandong Province, China
| | - Xiaomei Li
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Jianting Hu
- Shandong Pharmaceutical Academy, Shandong Provincial Key Laboratory of Chemical Drugs, Jinan, Shandong Province, China
| | - Xiaoyan Li
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
| | - Juan Xiao
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
| | - Yibo Dai
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
| | - Mingming He
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
| | - Xiaoli Liu
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| | - Yang Jiang
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| | - Ruodi Cui
- Department of Radiology, Qilu Children's Hospital of Shandong University, Jinan, Shandong Province, China
| | - Lihong Zhang
- Department of Pathology, Qilu Children's Hospital of Shandong University, Jinan, Shandong Province, China
| | - Juandong Wang
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| | - Ai Li
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| | - Fang Wang
- Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yuan Zhang
- Center of Evidence-Based Medicine, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Juan Xiao
- Center of Evidence-Based Medicine, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Wei Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Chengyun Zheng
- Department of hematology of the Second Hospital, Institute of Biotherapy for Hematological Malignancies, Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, Shandong Province, China
- Department of Hematology, Zhaoyuan Sorting-Yingcheng Hospital, Second Hospital of Shandong University, Yantai, Shandong Province, China
| |
Collapse
|
173
|
Abstract
PURPOSE OF REVIEW The advent of cell therapies, mainly based on the use of mesenchymal stromal cells (MSCs), represents a great step forward in the treatment of immune-mediated conditions. Here, we focus on those intestinal disorders wherein MSCs have been applied for immunotherapeutic purposes and whose results are available. RECENT FINDINGS By virtue of their ability to favour both tissue regeneration and immune tolerance, together with a substantial lack of immunogenicity, MSCs have gained huge attention in the last decade. Following abundant positive experimental data, a sizable number of clinical trials using MSCs as a new treatment in chronic inflammatory intestinal diseases were carried out with promising results and several are still ongoing. The main indication was refractory Crohn's disease wherein both feasibility and safety clearly emerged when treating the luminal phenotype with intravenous infusion/s, albeit no definitive conclusion on efficacy may be drawn. By contrast, the availability of robust demonstration also on the efficacy when treating the fistulizing phenotype through local injection/s of MSCs has led to approval of the marketing of an industrial preparation (darvadstrocel). SUMMARY Successful clinical implementation of this attractive option is hampered by a number of obstacles arising from methodology and regulation issues, which require the institution of interdisciplinary task forces before this cell therapy becomes a bedside reality.
Collapse
|
174
|
Mesenchymal Stem Cells Alleviate DHEA-Induced Polycystic Ovary Syndrome (PCOS) by Inhibiting Inflammation in Mice. Stem Cells Int 2019; 2019:9782373. [PMID: 31611920 PMCID: PMC6757294 DOI: 10.1155/2019/9782373] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in women of reproductive age. Chronic inflammation is considered to be the cause of ovarian dysfunction. Increasing evidence in animal studies and in preliminary clinical trials has demonstrated that MSCs possess immunomodulatory effects via their interaction with immune cells. However, their contribution to PCOS remains unclear. In this study, we showed that the administration of hUC-MSCs could efficiently improve the pathological changes of PCOS mice induced by dehydroepiandrosterone (DHEA), including ovarian histopathology and function. Moreover, we found that the administration of MSCs significantly downregulated the expression of proinflammatory factors (TNF-α, IL-1β, and IFN-γ) and fibrosis-related genes (CTGF) in ovarian and uterus tissues and affected the systemic inflammatory response. The percentage of peripheral neutrophils, M1 macrophages, and B cells was significantly reduced, while M2 macrophages and regulatory T cells (Tregs) were increased in hUC-MSC-treated mice. In the spleen, the percentage of neutrophils, M1 macrophages, IFN-γ+CD19+B cell, IFN-γ+CD4+T cells (Th1), and IL-17+CD4+T cells (Th17) was significantly decreased in hUC-MSC-treated mice. These results suggested that hUC-MSC treatment could alleviate ovarian dysfunction by inhibiting ovarian local and systemic inflammatory responses.
Collapse
|
175
|
Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 2019; 76:3323-3348. [PMID: 31055643 PMCID: PMC11105258 DOI: 10.1007/s00018-019-03125-1] [Citation(s) in RCA: 343] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are isolated from multiple biological tissues-adult bone marrow and adipose tissues and neonatal tissues such as umbilical cord and placenta. In vitro, MSCs show biological features of extensive proliferation ability and multipotency. Moreover, MSCs have trophic, homing/migration and immunosuppression functions that have been demonstrated both in vitro and in vivo. A number of clinical trials are using MSCs for therapeutic interventions in severe degenerative and/or inflammatory diseases, including Crohn's disease and graft-versus-host disease, alone or in combination with other drugs. MSCs are promising for therapeutic applications given the ease in obtaining them, their genetic stability, their poor immunogenicity and their curative properties for tissue repair and immunomodulation. The success of MSC therapy in degenerative and/or inflammatory diseases might depend on the robustness of the biological functions of MSCs, which should be linked to their therapeutic potency. Here, we outline the fundamental and advanced concepts of MSC biological features and underline the biological functions of MSCs in their basic and translational aspects in therapy for degenerative and/or inflammatory diseases.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Benoit Favier
- CEA, DRF-IBFJ, IDMIT, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang Occitanie, UMR 5273 CNRS, INSERM U1031, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Francois Jacob Institute, Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, IRSL, UMRS 976, Paris, France
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
176
|
Khan RS, Newsome PN. A Comparison of Phenotypic and Functional Properties of Mesenchymal Stromal Cells and Multipotent Adult Progenitor Cells. Front Immunol 2019; 10:1952. [PMID: 31555259 PMCID: PMC6724467 DOI: 10.3389/fimmu.2019.01952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022] Open
Abstract
Both Multipotent Adult Progenitor Cells and Mesenchymal Stromal Cells are bone-marrow derived, non-haematopoietic adherent cells, that are well-known for having immunomodulatory and pro-angiogenic properties, whilst being relatively non-immunogenic. However, they are phenotypically and functionally distinct cell types, which has implications for their efficacy in different settings. In this review we compare the phenotypic and functional properties of these two cell types, to help in determining which would be the superior cell type for different applications.
Collapse
Affiliation(s)
- Reenam S Khan
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
177
|
Laing AG, Riffo-Vasquez Y, Sharif-Paghaleh E, Lombardi G, Sharpe PT. Immune modulation by apoptotic dental pulp stem cells in vivo. Immunotherapy 2019; 10:201-211. [PMID: 29370720 DOI: 10.2217/imt-2017-0117] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) show considerable promise as a cellular immunotherapy for the treatment of a number of autoimmune and inflammatory disorders. However, the precise physiologically and therapeutically relevant mechanism(s) by which MSCs mediate immune modulation remains elusive. Dental pulp stem cells are a readily available source of MSCs that have been reported to show similar immune modulation in vitro as bone marrow MSCs. To test their potential in vivo, we used a clinically relevant humanized mouse model of GvHD in which only human T cells engraft. In this model, we found no effects on either T-cell proliferation, T-cell phenotype or disease progression. To determine if this lack of efficacy was related to a failure of engraftment or persistence of the cells, we used viability dependent radioactive cell tracking and showed that no cells were detectable after 24-h postinjection. Given the apparent failure of MSC to survive following intravenous injection, we hypothesized that their apoptosis may account for the widely reported therapeutic effect in numerous experimental models in vivo. To address this, we employed a well-established model of allergic airway inflammation to compare the efficacy of live and apoptotic MSCs in a fully immunocompetent model. In this model, both live and apoptotic dental pulp MSCs induced a robust immune suppressive reaction that was substantially greater with apoptotic cells. We propose that the mechanism of immune modulation following systemic application of MSCs is a result of cell entrapment and apoptosis occurring in the lungs.
Collapse
Affiliation(s)
- Adam G Laing
- MRC Centre for Transplantation, King's College London, London, UK, SE1 9RT.,Centre for Craniofacial & Regenerative Biology, King's College London, London, UK, SE1 9RT
| | - Yanira Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | | | - Giovanna Lombardi
- MRC Centre for Transplantation, King's College London, London, UK, SE1 9RT
| | - Paul T Sharpe
- MRC Centre for Transplantation, King's College London, London, UK, SE1 9RT.,Centre for Craniofacial & Regenerative Biology, King's College London, London, UK, SE1 9RT
| |
Collapse
|
178
|
Immune responses towards bioengineered tissues and strategies to control them. Curr Opin Organ Transplant 2019; 24:582-589. [PMID: 31385889 DOI: 10.1097/mot.0000000000000688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Research into development of artificial tissues and bioengineered organs to replace physiological functions of injured counterparts has highlighted a previously underestimated challenge for its clinical translatability: the immune response against biomaterials. Herein, we will provide an update and review current knowledge regarding this important barrier to regenerative medicine. RECENT FINDINGS Although a clear understanding of the immune reactivity against biomaterials remains elusive, accumulating evidence indicates that innate immune cells, primarily neutrophils and macrophages, play a key role in the initial phases of the immune response. More recently, data have shown that in later phases, T and B cells are also involved. The use of physicochemical modifications of biomaterials and cell-based strategies to modulate the host inflammatory response is being actively investigated for effective biomaterial integration. SUMMARY The immune response towards biomaterials and bioengineered organs plays a crucial role in determining their utility as transplantable grafts. Expanding our understanding of these responses is necessary for developing protolerogenic strategies and delivering on the ultimate promise of regenerative medicine.
Collapse
|
179
|
Zhang J, Ding H, Liu X, Sheng Y, Liu X, Jiang C. Dental Follicle Stem Cells: Tissue Engineering and Immunomodulation. Stem Cells Dev 2019; 28:986-994. [PMID: 30968740 DOI: 10.1089/scd.2019.0012] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jie Zhang
- Department of Orthodontics, the Affiliated Hospital of Qingdao University; School of Stomatology, Qingdao University, Qingdao, China
| | - Hong Ding
- Department of Orthodontics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinfeng Liu
- Department of Nuclear Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yunfei Sheng
- Department of Orthodontics, the Affiliated Hospital of Qingdao University; School of Stomatology, Qingdao University, Qingdao, China
| | - Xinqiang Liu
- Department of Orthodontics, the Affiliated Hospital of Qingdao University; School of Stomatology, Qingdao University, Qingdao, China
| | - Chunmiao Jiang
- Department of Orthodontics, the Affiliated Hospital of Qingdao University; School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
180
|
Zhu X, Wang Y, Jiang Q, Jiang H, Lu J, Wang Y, Kong Y, Chang Y, Xu L, Peng J, Hou M, Huang X, Zhang X. All- trans retinoic acid protects mesenchymal stem cells from immune thrombocytopenia by regulating the complement-interleukin-1β loop. Haematologica 2019; 104:1661-1675. [PMID: 30679324 PMCID: PMC6669169 DOI: 10.3324/haematol.2018.204446] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Enhanced peripheral complement activation has long been considered as one of the major pathogenic elements of immune thrombocytopenia. A dysfunctional bone marrow microenvironment, especially with regards to mesenchymal stem cells, has been observed in patients with immune thrombocytopenia. However, the potential role of the complement system in the dysfunctional bone marrow microenvironment remains poorly understood. In this study, bone marrow samples from patients with immune thrombocytopenia were divided into two groups based on whether or not complement components were deposited on the surfaces of their mesenchymal stem cells. The mesenchymal cells from the group with complement deposition were less numerous, dysfunctional, had a reduced capacity to proliferate, and showed increased apoptosis as well as abnormal secretion of interleukin-1β and C-X-C motif chemokine ligand 12. In vitro treatment with all-trans retinoic acid increased the number and improved the function of the complement-positive bone marrow mesenchymal stem cells by upregulating DNA hypermethylation of the interleukin-1β promoter. In vivo studies showed that all-trans retinoic acid could rescue the impaired mesenchymal stem cells to support the thrombopoietic niche in both patients with immune thrombocytopenia and a murine model of this disease. Taken together, these results indicate that impairment of mesenchymal stem cells, mediated by the complement-interleukin-1β loop, plays a role in the pathogenesis of immune thrombocytopenia. All-trans retinoic acid represents a promising therapeutic approach in patients with immune thrombocytopenia through its effect of repairing mesenchymal stem cell impairment.
Collapse
Affiliation(s)
- Xiaolu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yanan Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Hao Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Jin Lu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yazhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| |
Collapse
|
181
|
Das R, Roosloot R, van Pel M, Schepers K, Driessen M, Fibbe WE, de Bruijn JD, Roelofs H. Preparing for cell culture scale-out: establishing parity of bioreactor- and flask-expanded mesenchymal stromal cell cultures. J Transl Med 2019; 17:241. [PMID: 31340829 PMCID: PMC6657181 DOI: 10.1186/s12967-019-1989-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Cell-based therapies have the potential to become treatment options for many diseases, but efficient scale-out of these therapies has proven to be a major hurdle. Bioreactors can be used to overcome this hurdle, but changing the culture method can introduce unwanted changes to the cell product. Therefore, it is important to establish parity between products generated using traditional methods versus those generated using a bioreactor. Methods Mesenchymal stromal cells (MSCs) are cultured in parallel using either traditional culture flasks, spinner vessels or a new bioreactor system. To investigate parity between the cells obtained from different methods, harvested cells are compared in terms of yield, phenotype and functionality. Results Bioreactor-based expansion yielded high cell numbers (222–510 million cells). Highest cell expansion was observed upon culture in flasks [average 5.0 population doublings (PDL)], followed by bioreactor (4.0 PDL) and spinner flasks (3.3 PDL). Flow cytometry confirmed MSC identity (CD73+, CD90+ and CD105+) and lack of contaminating hematopoietic cell populations. Cultured MSCs did not display genetic aberrations and no difference in differentiation and immunomodulatory capacity was observed between culture conditions. The response to IFNγ stimulation was similar for cells obtained from all culture conditions, as was the capacity to inhibit T cell proliferation. Conclusions The new bioreactor technology can be used to culture large amounts of cells with characteristics equivalent to those cultured using traditional, flask based, methods. Electronic supplementary material The online version of this article (10.1186/s12967-019-1989-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruud Das
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.
| | - Rens Roosloot
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Melissa van Pel
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Koen Schepers
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marijn Driessen
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Willem E Fibbe
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Joost Dick de Bruijn
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.,Twente University, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Helene Roelofs
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
182
|
Taechangam N, Iyer SS, Walker NJ, Arzi B, Borjesson DL. Mechanisms utilized by feline adipose-derived mesenchymal stem cells to inhibit T lymphocyte proliferation. Stem Cell Res Ther 2019; 10:188. [PMID: 31238978 PMCID: PMC6593543 DOI: 10.1186/s13287-019-1300-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/20/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Feline adipose-derived mesenchymal stem cells (ASCs) have been successfully used in clinical trials for the treatment of immune-mediated diseases with T cell dysregulation. However, the immunomodulatory pathways utilized by feline ASCs to suppress T cell activation have not been fully determined. We investigated the mechanisms used by feline ASCs to inhibit T cell proliferation, including the soluble factors and the cell-cell contact ligands responsible for ASC-T cell interaction. METHODS The immunomodulatory activity of feline ASCs was evaluated via cell cycle analysis and in vitro mixed leukocyte reaction using specific immunomodulatory inhibitors. Cell-cell interactions were assessed with static adhesion assays, also with inhibitors. RESULTS Feline ASCs decrease T cell proliferation by causing cell cycle arrest in G0-G1. Blocking prostaglandin (PGE2), but not IDO, partially restored lymphocyte proliferation. Although PDL-1 and CD137L are both expressed on activated feline ASCs, only the interaction of intercellular adhesion molecule 1 (ICAM-1, CD54) with its ligand, lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), was responsible for ASC-T cell adhesion. Blocking this interaction reduced cell-cell adhesion and mediator (IFN-γ) secretion and signaling. CONCLUSIONS Feline ASCs utilize PGE2 and ICAM-1/LFA-1 ligand interaction to inhibit T cell proliferation with a resultant cell cycle arrest in G0-G1. These data further elucidate the mechanisms by which feline ASCs interact with T cells, help define appropriate T cell-mediated disease targets in cats that may be amenable to ASC therapy, and may also inform potential translational models for human diseases.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Smita S Iyer
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, University of California, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA. .,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
183
|
Yu-Taeger L, Stricker-Shaver J, Arnold K, Bambynek-Dziuk P, Novati A, Singer E, Lourhmati A, Fabian C, Magg J, Riess O, Schwab M, Stolzing A, Danielyan L, Nguyen HHP. Intranasal Administration of Mesenchymal Stem Cells Ameliorates the Abnormal Dopamine Transmission System and Inflammatory Reaction in the R6/2 Mouse Model of Huntington Disease. Cells 2019; 8:E595. [PMID: 31208073 PMCID: PMC6628278 DOI: 10.3390/cells8060595] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022] Open
Abstract
Intrastriatal administration of mesenchymal stem cells (MSCs) has shown beneficial effects in rodent models of Huntington disease (HD). However, the invasive nature of surgical procedure and its potential to trigger the host immune response may limit its clinical use. Hence, we sought to evaluate the non-invasive intranasal administration (INA) of MSC delivery as an effective alternative route in HD. GFP-expressing MSCs derived from bone marrow were intranasally administered to 4-week-old R6/2 HD transgenic mice. MSCs were detected in the olfactory bulb, midbrain and striatum five days post-delivery. Compared to phosphate-buffered saline (PBS)-treated littermates, MSC-treated R6/2 mice showed an increased survival rate and attenuated circadian activity disruption assessed by locomotor activity. MSCs increased the protein expression of DARPP-32 and tyrosine hydroxylase (TH) and downregulated gene expression of inflammatory modulators in the brain 7.5 weeks after INA. While vehicle treated R6/2 mice displayed decreased Iba1 expression and altered microglial morphology in comparison to the wild type littermates, MSCs restored both, Iba1 level and the thickness of microglial processes in the striatum of R6/2 mice. Our results demonstrate significantly ameliorated phenotypes of R6/2 mice after MSCs administration via INA, suggesting this method as an effective delivering route of cells to the brain for HD therapy.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Janice Stricker-Shaver
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Katrin Arnold
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Patrycja Bambynek-Dziuk
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Arianna Novati
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Elisabeth Singer
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Ali Lourhmati
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
| | - Claire Fabian
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Janine Magg
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Matthias Schwab
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, D-70376 Stuttgart, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Alexandra Stolzing
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Hoa Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany.
- Departments of Medical Chemistry and Biochemistry, Yerevan State Medical University, 0025 Yerevan, Armenia.
| |
Collapse
|
184
|
The Immunomodulatory Potential of Wharton's Jelly Mesenchymal Stem/Stromal Cells. Stem Cells Int 2019; 2019:3548917. [PMID: 31281372 PMCID: PMC6594275 DOI: 10.1155/2019/3548917] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/01/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022] Open
Abstract
The benefits attributed to mesenchymal stem/stromal cells (MSC) in cell therapy applications are mainly attributed to the secretion of factors, which exhibit immunomodulatory and anti-inflammatory effects and stimulate angiogenesis. Despite the desirable features such as high proliferation levels, multipotency, and immune response regulation, there are important variables that must be considered. Although presenting similar morphological aspects, MSC collected from different tissues can form heterogeneous cellular populations and, therefore, manifest functional differences. Thus, the source of MSC should be a factor to be considered in the development of novel therapies. The following text presents an updated review of recent research outcomes related to Wharton's jelly mesenchymal stem/stromal cells (WJ-MSC), harvested from umbilical cords and considered novel and potential candidates for the development of cell-based approaches. This text highlights information on how WJ-MSC affect immune responses in comparison with other sources of MSC.
Collapse
|
185
|
Podestà MA, Remuzzi G, Casiraghi F. Mesenchymal Stromal Cells for Transplant Tolerance. Front Immunol 2019; 10:1287. [PMID: 31231393 PMCID: PMC6559333 DOI: 10.3389/fimmu.2019.01287] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/21/2019] [Indexed: 12/18/2022] Open
Abstract
In solid organ transplantation lifelong immunosuppression exposes transplant recipients to life-threatening complications, such as infections and malignancies, and to severe side effects. Cellular therapy with mesenchymal stromal cells (MSC) has recently emerged as a promising strategy to regulate anti-donor immune responses, allowing immunosuppressive drug minimization and tolerance induction. In this review we summarize preclinical data on MSC in solid organ transplant models, focusing on potential mechanisms of action of MSC, including down-regulation of effector T-cell response and activation of regulatory pathways. We will also provide an overview of available data on safety and feasibility of MSC therapy in solid organ transplant patients, highlighting the issues that still need to be addressed before establishing MSC as a safe and effective tolerogenic cell therapy in transplantation.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.,Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Remuzzi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Federica Casiraghi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
186
|
Zhang Q, Li Q, Zhu J, Guo H, Zhai Q, Li B, Jin Y, He X, Jin F. Comparison of therapeutic effects of different mesenchymal stem cells on rheumatoid arthritis in mice. PeerJ 2019; 7:e7023. [PMID: 31198641 PMCID: PMC6553443 DOI: 10.7717/peerj.7023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic and nonspecific autoimmune disease, which leads to joint destruction and deformity. To investigate the potential of human mesenchymal stem cells (MSCs) as a new therapeutic strategy for patients with RA, we compared the therapeutic effects of bone marrow derived MSCs (BMSCs), umbilical cord derived MSCs (UCs), and stem cells derived from human exfoliated deciduous teeth (SHED) on collagen-induced arthritis (CIA) in mice. Methods A total of 24 DBA/1 mice were infused with type II collagen to induce RA in the experimental model. MSC-treated mice were infused with UCs, BMSCs, and SHED, respectively. Bone erosion and joint destruction were measured by micro-computed tomographic (micro-CT) analysis and hematoxylin and eosin staining. The levels of tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β) were measured by immunohistochemistry and Enzyme-Linked Immunosorbent Assay (ELISA). Results Systemic delivery of MSCs significantly improved the severity of the symptoms related to CIA to greater extent compared with the untreated control group. Micro-CT revealed reduced bone erosions in the metatarsophalangeal joints upon treatment with MSCs. Additionally, according to histologic evaluation, reduced synovitis and articular destruction were observed in MSC-treated groups. The levels of TNF-α and IL-1β in the serum and joints decreased with treatment by MSCs. Conclusion Our findings suggest that systemic infusion of UCs, BMSCs, and SHED may significantly alleviate the effects of RA. The therapeutic effect of BMSCs was greater than that of SHED, while the UCs were shown to have the best therapeutic effect on CIA mice. In conclusion, compared with BMSCs and SHED, UCs may be a more suitable source of MSCs for the treatment of patients with RA.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Qihong Li
- Department of Stomatology, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jun Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Hao Guo
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Qiming Zhai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Xiaoning He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Fang Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical Universit, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
187
|
Mesenchymal stem cells immunomodulation: The road to IFN-γ licensing and the path ahead. Cytokine Growth Factor Rev 2019; 47:32-42. [DOI: 10.1016/j.cytogfr.2019.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
|
188
|
Andrzejewska A, Lukomska B, Janowski M. Concise Review: Mesenchymal Stem Cells: From Roots to Boost. Stem Cells 2019; 37:855-864. [PMID: 30977255 DOI: 10.1002/stem.3016] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/25/2019] [Accepted: 03/31/2019] [Indexed: 12/13/2022]
Abstract
It was shown as long as half a century ago that bone marrow is a source of not only hematopoietic stem cells, but also stem cells of mesenchymal tissues. Then the term "mesenchymal stem cells" (MSCs) was coined in the early 1990s, and more than a decade later, the criteria for defining MSCs have been released by the International Society for Cellular Therapy. The easy derivation from a variety of fetal and adult tissues and undemanding cell culture conditions made MSCs an attractive research object. It was followed by the avalanche of reports from preclinical studies on potentially therapeutic properties of MSCs, such as immunomodulation, trophic support and capability for a spontaneous differentiation into connective tissue cells, and differentiation into the majority of cell types upon specific inductive conditions. Although ontogenesis, niche, and heterogeneity of MSCs are still under investigation, there is a rapid boost of attempts at clinical applications of MSCs, especially for a flood of civilization-driven conditions in so quickly aging societies, not only in the developed countries, but also in the populous developing world. The fields of regenerative medicine and oncology are particularly extensively addressed by MSC applications, in part due to the paucity of traditional therapeutic options for these highly demanding and costly conditions. There are currently almost 1,000 clinical trials registered worldwide at ClinicalTrials.gov, and it seems that we are starting to witness the snowball effect with MSCs becoming a powerful global industry; however, the spectacular effects of MSCs in the clinic still need to be shown. Stem Cells 2019;37:855-864.
Collapse
Affiliation(s)
- Anna Andrzejewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
189
|
Razmkhah M, Abtahi S, Ghaderi A. Mesenchymal Stem Cells, Immune Cells and Tumor Cells Crosstalk: A Sinister Triangle in the Tumor Microenvironment. Curr Stem Cell Res Ther 2019; 14:43-51. [PMID: 30112998 DOI: 10.2174/1574888x13666180816114809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal Stem Cells [MSCs] are a heterogeneous population of fibroblast-like cells which maintain self-renewability and pluripotency. Many studies have demonstrated the immunomodulatory effects of MSCs on the innate and adaptive immune cells. As a result of interactions with tumor cells, microenvironment and immune-stimulating milieu, MSCs contribute to tumor progression by several mechanisms, including sustained proliferative signal in cancer stem cells [CSCs], inhibition of tumor cell apoptosis, transition to tumor-associated fibroblasts [TAFs], promotion of angiogenesis, stimulation of epithelial-mesenchymal transition [EMT], suppression of immune responses, and consequential promotion of tumor metastasis. Here, we present an overview of the latest findings on Janusfaced roles that MSCs play in the tumor microenvironment [TME], with a concise focus on innate and adaptive immune responses.
Collapse
Affiliation(s)
- Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shabnam Abtahi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
190
|
Hu C, Zhao L, Wu D, Li L. Modulating autophagy in mesenchymal stem cells effectively protects against hypoxia- or ischemia-induced injury. Stem Cell Res Ther 2019; 10:120. [PMID: 30995935 PMCID: PMC6471960 DOI: 10.1186/s13287-019-1225-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In mammals, a basal level of autophagy, a self-eating cellular process, degrades cytosolic proteins and subcellular organelles in lysosomes to provide energy, recycles the cytoplasmic components, and regenerates cellular building blocks; thus, autophagy maintains cellular and tissue homeostasis in all eukaryotic cells. In general, adaptive autophagy increases when cells confront stressful conditions to improve the survival rate of the cells, while destructive autophagy is activated when the cellular stress is not manageable and elicits the regenerative capacity. Hypoxia-reoxygenation (H/R) injury and ischemia-reperfusion (I/R) injury initiate excessive autophagy and endoplasmic reticulum (ER) stress and consequently induce a string of damage in mammalian tissues or organs. Mesenchymal stem cell (MSC)-based therapy has yielded promising results in repairing H/R- or I/R-induced injury in various tissues. However, MSC transplantation in vivo must overcome the barriers including the low survival rate of transplanted stem cells, limited targeting capacity, and low grafting potency; therefore, much effort is needed to increase the survival and activity of MSCs in vivo. Modulating autophagy regulates the stemness and the anti-oxidative stress, anti-apoptosis, and pro-survival capacity of MSCs and can be applied to MSC-based therapy for repairing H/R- or I/R-induced cellular or tissue injury.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lingfei Zhao
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Daxian Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
191
|
Gao WH, Gao HY, Li YT, Huang PP. Effectiveness of umbilical cord mesenchymal stem cells in patients with critical limb ischemia. Med Clin (Barc) 2019; 153:341-346. [PMID: 30926157 DOI: 10.1016/j.medcli.2019.01.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION AND OBJECTIVE Transplantation of umbilical cord mesenchymal stem cells (UC-MSCs) has been shown to be effective in treating critical limb ischemia (CLI). However, the mechanism of MSCs-mediated improvements, especially on the immune-inflammatory aspects of this disease, is still unknown. In this study, we investigated the changes in T-lymphocyte subpopulations and inflammatory mediators (such as IL-6, IL-10 and TNF-α) in PBMCs from CLI patients after UC-MSCs treatment and correlation between inflammatory mediators and EPCs. PATIENTS AND METHODS 8 patients received UC-MSCs transplantation. Before the treatment, at 24h and 1 month thereafter, peripheral blood samples were collected from 8 patients and 8 healthy volunteers. Patients were evaluated for changes in IL-6, IL-10, TNF-α and levels of circulating EPCs. RESULTS TNF-α and IL-6 serum levels increased at 24h (p=0.017, p=0.099) after treatment and then decreased at 1 month (p=0.031, p=0.072) compared with those before treatment. The percentages of CD3+T, CD3+CD4+T-lymphocytes and NK cells decreased significantly after UC-MSCs treatment (p=0.002, p=0.012 and p=0.029, respectively). TNF-α (r=-0.602, p=0.038) was shown to be inversely correlated with the number of circulating EPCs. CONCLUSIONS This study demonstrates that UC-MSCs have anti-inflammatory and immunomodulation properties in CLI and suggests that UC-MSCs promote healing of non-healing wounds.
Collapse
Affiliation(s)
- Wen-Hui Gao
- Institute of Hematology, General Medical Center, Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hong-Ye Gao
- Institute of Hematology, General Medical Center, Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yue-Tong Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ping-Ping Huang
- Institute of Hematology, General Medical Center, Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| |
Collapse
|
192
|
Laing AG, Fanelli G, Ramirez-Valdez A, Lechler RI, Lombardi G, Sharpe PT. Mesenchymal stem cells inhibit T-cell function through conserved induction of cellular stress. PLoS One 2019; 14:e0213170. [PMID: 30870462 PMCID: PMC6417714 DOI: 10.1371/journal.pone.0213170] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 02/15/2019] [Indexed: 12/17/2022] Open
Abstract
The physiological role of mesenchymal stem cells (MSCs) is to provide a source of cells to replace mesenchymal-derivatives in stromal tissues with high cell turnover or following stromal tissue damage to elicit repair. Human MSCs have been shown to suppress in vitro T-cell responses via a number of mechanisms including indoleamine 2,3-dioxygenase (IDO). This immunomodulatory capacity is likely to be related to their in vivo function in tissue repair where local, transient suppression of immune responses would benefit differentiation. Further understanding of the impact of locally modulated immune responses by MSCs is hampered by evidence that IDO is not produced or utilized by mouse MSCs. In this study, we demonstrate that IDO-mediated tryptophan starvation triggered by human MSCs inhibits T-cell activation and proliferation through induction of cellular stress. Significantly, we show that despite utilizing different means, immunomodulation of murine T-cells also involves cellular stress and thus is a common strategy of immunoregulation conserved between mouse and humans.
Collapse
Affiliation(s)
- Adam G. Laing
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King’s College London, United Kingdom
| | - Giorgia Fanelli
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, United Kingdom
| | - Andrei Ramirez-Valdez
- Centre for Craniofacial and Regenerative Biology, King’s College London, United Kingdom
| | - Robert I. Lechler
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, United Kingdom
| | - Giovanna Lombardi
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, United Kingdom
| | - Paul T. Sharpe
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King’s College London, United Kingdom
| |
Collapse
|
193
|
Thaweesapphithak S, Tantrawatpan C, Kheolamai P, Tantikanlayaporn D, Roytrakul S, Manochantr S. Human serum enhances the proliferative capacity and immunomodulatory property of MSCs derived from human placenta and umbilical cord. Stem Cell Res Ther 2019; 10:79. [PMID: 30845980 PMCID: PMC6407186 DOI: 10.1186/s13287-019-1175-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/07/2019] [Accepted: 02/12/2019] [Indexed: 12/29/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are considered potential candidates that hold great promise in the treatment of immune-related diseases. For therapeutic applications, it is necessary to isolate and expand MSCs with procedures complying with good manufacturing practice (GMP). Recent studies reported the use of human serum (HS) instead of fetal bovine serum (FBS) for the expansion of bone marrow-derived MSCs. Nevertheless, there are only limited data on HS as an alternative to FBS for the isolation and expansion of umbilical (UC-MSCs) and placenta-derived MSCs (PL-MSCs). In this study, we evaluate the effect of HS compared to FBS on the proliferative and immunosuppressive capacities of these MSCs. Methods PL-MSCs and UC-MSCs were isolated and cultured in HS- or FBS-supplemented media. The MSC characteristics, including morphology, immunophenotype, and differentiation ability, were verified. The proliferative and immunosuppressive capacities were also examined. In addition, the proliferative-enhancing factors in both sera were explored using proteomic analysis. Results PL-MSCs and UC-MSCs proliferated faster in HS-supplemented medium than in equivalent levels of FBS-supplemented medium. Adipogenic and osteogenic differentiations occurred at nearly identical levels in HS- and FBS-supplemented media. Interestingly, MSCs cultured in HS-supplemented medium had a similar immunosuppressive effect as MSCs cultured in FBS-supplemented medium. Proteomic analysis revealed that Con-A binding glycoproteins with a molecular weight > 100 kDa in FBS could significantly enhance MSC proliferation. In contrast, the proliferative enhancing factors in HS were found in the Con-A non-binding fraction and WGA binding fraction with a molecular weight > 100 kDa. Conclusions Taken together, our results suggest applications for the use of HS instead of FBS for the isolation and expansion of PL-MSCs and UC-MSCs for cell therapy in the future. Furthermore, this study identifies factors in HS that are responsible for its proliferative and immunosuppressive effects and might thus lead to the establishment of GMPs for the therapeutic use of MSCs.
Collapse
Affiliation(s)
- Sermporn Thaweesapphithak
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Chairat Tantrawatpan
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Pakpoom Kheolamai
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Sittiruk Roytrakul
- Genome Institute, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sirikul Manochantr
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand. .,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
194
|
Shen Y, Xue C, Li X, Ba L, Gu J, Sun Z, Han Q, Zhao RC. Effects of Gastric Cancer Cell-Derived Exosomes on the Immune Regulation of Mesenchymal Stem Cells by the NF-kB Signaling Pathway. Stem Cells Dev 2019; 28:464-476. [PMID: 30717632 DOI: 10.1089/scd.2018.0125] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are important components of the tumor microenvironment, which play an important role in tumor development. Exosomes derived from tumor cells can affect the biological characteristics of MSCs. Our study examined the effects of exosomes derived from gastric cancer cells on MSC immunomodulatory functions. Exosomes were extracted from gastric cancer cell line AGS (AGS-Exos) and cultured with MSCs. MSCs were then cocultured with both human peripheral blood mononuclear cells and macrophages [phorbol-12-myristate-13-acetate (PMA)-stimulated THP1 cells]. The activation levels of T cells and macrophages were detected by flow cytometry and real-time quantitative polymerase chain reaction (RT-PCR). Changes in the MSC signaling pathway after AGS-Exos stimulation were studied using RNA Chip, and the molecular mechanisms of functional change in MSCs were studied by inhibiting the signaling pathway. MSCs treated with AGS-Exos could promote macrophage phagocytosis and upregulate the secretion of proinflammatory factor, and promote the activation of CD69 and CD25 on the surface of T cells. RNA Chip results indicated the abnormal activation of the NF-kB signaling pathway in MSCs after AGS-Exos stimulation, and this was verified by the identification of key proteins in the pathway using western blot analysis. After NF-kB signaling pathway inhibition, the effect of MSCs stimulated by AGS-Exos on T cells and macrophages was markedly weakened. Therefore, AGS-Exos affected the immunomodulation function of MSCs through the NF-kB signaling pathway, which enhanced the ability of MSCs to activate immune cells, maintain the inflammatory environment, and support tumor growth.
Collapse
Affiliation(s)
- Yamei Shen
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| | - Chunling Xue
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| | - Xuechun Li
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| | - Li Ba
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| | - Junjie Gu
- 2 Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhao Sun
- 2 Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Qin Han
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| | - Robert Chunhua Zhao
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No. BZO381), Beijing, People's Republic of China
| |
Collapse
|
195
|
Erpicum P, Weekers L, Detry O, Bonvoisin C, Delbouille MH, Grégoire C, Baudoux E, Briquet A, Lechanteur C, Maggipinto G, Somja J, Pottel H, Baron F, Jouret F, Beguin Y. Infusion of third-party mesenchymal stromal cells after kidney transplantation: a phase I-II, open-label, clinical study. Kidney Int 2019; 95:693-707. [DOI: 10.1016/j.kint.2018.08.046] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/25/2018] [Accepted: 08/23/2018] [Indexed: 02/08/2023]
|
196
|
Gugjoo MB, Amarpal A, Sharma GT. Mesenchymal stem cell basic research and applications in dog medicine. J Cell Physiol 2019; 234:16779-16811. [PMID: 30790282 DOI: 10.1002/jcp.28348] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
The stem cells, owing to their special characteristics like self-renewal, multiplication, homing, immunomodulation, anti-inflammatory, and dedifferentiation are considered to carry an "all-in-one-solution" for diverse clinical problems. However, the limited understanding of cellular physiology currently limits their definitive therapeutic use. Among various stem cell types, currently mesenchymal stem cells are extensively studied for dog clinical applications owing to their readily available sources, easy harvesting, and ability to differentiate both into mesodermal, as well as extramesodermal tissues. The isolated, culture expanded, and characterized cells have been applied both at preclinical as well as clinical settings in dogs with variable but mostly positive results. The results, though positive, are currently inconclusive and demands further intensive research on the properties and their dependence on the applications. Further, numerous clinical conditions of dog resemble to that of human counterparts and thus, if proved rewarding in the former may act as basis of therapy for the latter. The current review throws some light on dog mesenchymal stem cell properties and their potential therapeutic applications.
Collapse
Affiliation(s)
- Mudasir Bashir Gugjoo
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Jammu and Kashmir, India
| | - Amarpal Amarpal
- Division of Surgery, Indian Veterinary Research Institute, Izatnagar, India
| | - Gutulla Taru Sharma
- Division of Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
197
|
Poglajen G, Gregoric ID, Radovancevic R, Vrtovec B. Stem Cell and Left Ventricular Assist Device Combination Therapy. Circ Heart Fail 2019; 12:e005454. [PMID: 30759999 DOI: 10.1161/circheartfailure.118.005454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ventricular assist device (VAD) technology has evolved significantly over the past decades and currently represents one of the most important treatment strategies for patients with advanced chronic heart failure. There is increasing evidence that in selected patients undergoing long-term VAD support, improvement of myocardial structure and function may occur. However, there seems to be a significant discrepancy between structural and functional recovery of the failing myocardium, as only a small fraction of VAD-supported patients demonstrate reverse structural remodeling and eventually reach clinically significant and stable, functional improvement. More recently, cell therapy has gained a growing interest in the heart failure community because of its potential to augment reverse remodeling of the failing myocardium. Although theoretically the combination of long-term VAD support and cell therapy may offer significant advantages over using these therapeutic modalities separately, it remains largely unexplored. This review aims to summarize the current state of the art of the effects of VAD support and cell therapy on the reverse remodeling of the failing myocardium and to discuss the rationale for using a combined treatment strategy to further promote myocardial recovery in patients with advanced chronic heart failure.
Collapse
Affiliation(s)
- Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia (G.P., B.V.).,Department of Advanced Cardiopulmonary Therapies and Transplantation, Center for Advanced Heart Failure, University of Texas Health Science Center at Houston (G.P., I.D.G., R.R.)
| | - Igor D Gregoric
- Department of Advanced Cardiopulmonary Therapies and Transplantation, Center for Advanced Heart Failure, University of Texas Health Science Center at Houston (G.P., I.D.G., R.R.)
| | - Rajko Radovancevic
- Department of Advanced Cardiopulmonary Therapies and Transplantation, Center for Advanced Heart Failure, University of Texas Health Science Center at Houston (G.P., I.D.G., R.R.)
| | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia (G.P., B.V.)
| |
Collapse
|
198
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:jcm8020249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
199
|
Bertheuil N, Chaput B, Ménard C, Varin A, Laloze J, Watier E, Tarte K. Adipose mesenchymal stromal cells: Definition, immunomodulatory properties, mechanical isolation and interest for plastic surgery. ANN CHIR PLAST ESTH 2019; 64:1-10. [DOI: 10.1016/j.anplas.2018.07.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022]
|
200
|
Mesenchymal stem cells for inflammatory airway disorders: promises and challenges. Biosci Rep 2019; 39:BSR20182160. [PMID: 30610158 PMCID: PMC6356012 DOI: 10.1042/bsr20182160] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
The regenerative and immunomodulatory characteristics of mesenchymal stem cells (MSCs) make them attractive in the treatment of many diseases. Although they have shown promising preclinical studies of immunomodulation and paracrine effects in inflammatory airway disorders and other lung diseases, there are still challenges that have to be overcome before MSCs can be safely, effectively, and routinely applied in the clinical setting. A good understanding of the roles and mechanisms of the MSC immunomodulatory effects will benefit the application of MSC-based clinical therapy. In this review, we summarize the promises and challenges of the preclinical and clinical trials of MSC therapies, aiming to better understand the role that MSCs play in attempt to treat inflammatory airway disorders.
Collapse
|