101
|
Massa M, Croce S, Campanelli R, Abbà C, Lenta E, Valsecchi C, Avanzini MA. Clinical Applications of Mesenchymal Stem/Stromal Cell Derived Extracellular Vesicles: Therapeutic Potential of an Acellular Product. Diagnostics (Basel) 2020; 10:diagnostics10120999. [PMID: 33255416 PMCID: PMC7760121 DOI: 10.3390/diagnostics10120999] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decade, the secreting activity of mesenchymal stem/stromal cells (MSCs) has been widely investigated, due to its possible therapeutic role. In fact, MSCs release extracellular vesicles (EVs) containing relevant biomolecules such as mRNAs, microRNAs, bioactive lipids, and signaling receptors, able to restore physiological conditions where regenerative or anti-inflammatory actions are needed. An actual advantage would come from the therapeutic use of EVs with respect to MSCs, avoiding the possible immune rejection, the lung entrapment, improving the safety, and allowing the crossing of biological barriers. A number of concerns still have to be solved regarding the mechanisms determining the beneficial effect of MSC-EVs, the possible alteration of their properties as a consequence of the isolation/purification methods, and/or the best approach for a large-scale production for clinical use. Most of the preclinical studies have been successful, reporting for MSC-EVs a protecting role in acute kidney injury following ischemia reperfusion, a potent anti-inflammatory and anti-fibrotic effects by reducing disease associated inflammation and fibrosis in lung and liver, and the modulation of both innate and adaptive immune responses in graft versus host disease (GVHD) as well as autoimmune diseases. However, the translation of MSC-EVs to the clinical stage is still at the initial phase. Herein, we discuss the therapeutic potential of an acellular product such as MSC derived EVs (MSC-EVs) in acute and chronic pathologies.
Collapse
Affiliation(s)
- Margherita Massa
- Biochemistry, Biotechnology and Advanced Diagnostics Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (M.M.); (C.A.)
| | - Stefania Croce
- General Surgery Department, Fondazione IRCCS Policlinico S. Matteo, Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Rita Campanelli
- Center for the Study of Myelofibrosis, Biochemistry, Biotechnology and Advanced Diagnostics Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Carlotta Abbà
- Biochemistry, Biotechnology and Advanced Diagnostics Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (M.M.); (C.A.)
| | - Elisa Lenta
- Cell Factory, Pediatric Hematology Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Chiara Valsecchi
- Immunology and Transplantation Laboratory, Cell Factory, Pediatric Hematology Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Maria Antonietta Avanzini
- Immunology and Transplantation Laboratory, Cell Factory, Pediatric Hematology Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
- Correspondence:
| |
Collapse
|
102
|
Therapeutic Potential of Mesenchymal Stem Cells and Their Secretome in the Treatment of SARS-CoV-2-Induced Acute Respiratory Distress Syndrome. ACTA ACUST UNITED AC 2020; 2020:1939768. [PMID: 33274176 PMCID: PMC7678745 DOI: 10.1155/2020/1939768] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/06/2020] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent responsible for the development of a new coronavirus disease (COVID-19), is a highly transmittable virus which, in just ten months, infected more than 40 million people in 214 countries worldwide. After inhalation, aerosols containing SARS-CoV-2 penetrate to the depths of the lungs and cause severe pneumonia, alveolar injury, and life-threatening acute respiratory distress syndrome (ARDS). Since there are no specific drugs or vaccines available to cure or prevent COVID-19 infection and COVID-19-related ARDS, a new therapeutic agent which will support oxygen supply and, at the same time, efficiently alleviate SARS-CoV-2-induced lung inflammation is urgently needed. Due to their potent immuno- and angiomodulatory characteristics, mesenchymal stem cells (MSCs) may increase oxygen supply in the lungs and may efficiently alleviate ongoing lung inflammation, including SARS-CoV-2-induced ARDS. In this review article, we described molecular mechanisms that are responsible for MSC-based modulation of immune cells which play a pathogenic role in the development of SARS-CoV-2-induced ARDS and we provided a brief outline of already conducted and ongoing clinical studies that increase our understanding about the therapeutic potential of MSCs and their secretome in the therapy of COVID-19-related ARDS.
Collapse
|
103
|
Wise RM, Harrison MAA, Sullivan BN, Al-Ghadban S, Aleman SJ, Vinluan AT, Monaco ER, Donato UM, Pursell IA, Bunnell BA. Short-Term Rapamycin Preconditioning Diminishes Therapeutic Efficacy of Human Adipose-Derived Stem Cells in a Murine Model of Multiple Sclerosis. Cells 2020; 9:E2218. [PMID: 33008073 PMCID: PMC7600854 DOI: 10.3390/cells9102218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 01/22/2023] Open
Abstract
Human adipose-derived stem cells (ASCs) show immense promise for treating inflammatory diseases, attributed primarily to their potent paracrine signaling. Previous investigations demonstrated that short-term Rapamycin preconditioning of bone marrow-derived stem cells (BMSCs) elevated secretion of prostaglandin E2, a pleiotropic molecule with therapeutic effects in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS), and enhanced immunosuppressive capacity in vitro. However, this has yet to be examined in ASCs. The present study examined the therapeutic potential of short-term Rapamycin-preconditioned ASCs in the EAE model. Animals were treated at peak disease with control ASCs (EAE-ASCs), Rapa-preconditioned ASCs (EAE-Rapa-ASCs), or vehicle control (EAE). Results show that EAE-ASCs improved clinical disease scores and elevated intact myelin compared to both EAE and EAE-Rapa-ASC animals. These results correlated with augmented CD4+ T helper (Th) and T regulatory (Treg) cell populations in the spinal cord, and increased gene expression of interleukin-10 (IL-10), an anti-inflammatory cytokine. Conversely, EAE-Rapa-ASC mice showed no improvement in clinical disease scores, reduced myelin levels, and significantly less Th and Treg cells in the spinal cord. These findings suggest that short-term Rapamycin preconditioning reduces the therapeutic efficacy of ASCs when applied to late-stage EAE.
Collapse
Affiliation(s)
- Rachel M. Wise
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
| | - Mark A. A. Harrison
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
| | - Brianne N. Sullivan
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
| | - Sara Al-Ghadban
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Sarah J. Aleman
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
| | - Amber T. Vinluan
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
| | - Emily R. Monaco
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
| | - Umberto M. Donato
- Neuroscience Program, Tulane Brain Institute, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA; (R.M.W.); (M.A.A.H.); (B.N.S.); (S.J.A.); (A.T.V.); (E.R.M.); (U.M.D.)
| | - India A. Pursell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
| | - Bruce A. Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.A.-G.); (I.A.P.)
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
104
|
Mirershadi F, Ahmadi M, Rezabakhsh A, Rajabi H, Rahbarghazi R, Keyhanmanesh R. Unraveling the therapeutic effects of mesenchymal stem cells in asthma. Stem Cell Res Ther 2020; 11:400. [PMID: 32933587 PMCID: PMC7493154 DOI: 10.1186/s13287-020-01921-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
Asthma is a chronic inflammatory disease associated with airway hyper-responsiveness, chronic inflammatory response, and excessive structural remodeling. The current therapeutic strategies in asthmatic patients are based on controlling the activity of type 2 T helper lymphocytes in the pulmonary tissue. However, most of the available therapies are symptomatic and expensive and with diverse side outcomes in which the interruption of these modalities contributes to the relapse of asthmatic symptoms. Up to date, different reports highlighted the advantages and beneficial outcomes regarding the transplantation of different stem cell sources, and relevant products from for the diseases' alleviation and restoration of injured sites. However, efforts to better understand by which these cells elicit therapeutic effects are already underway. The precise understanding of these mechanisms will help us to translate stem cells into the clinical setting. In this review article, we described current knowledge and future perspectives related to the therapeutic application of stem cell-based therapy in animal models of asthma, with emphasis on the underlying therapeutic mechanisms.
Collapse
Affiliation(s)
- Fatemeh Mirershadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran.,Department of Physiology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Rajabi
- Koc University Research Center for Translational Medicine (KUTTAM), Koc University School of Medicine, Istanbul, Turkey.,Department of Pulmonary Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51548-53431, Iran.
| | - Rana Keyhanmanesh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Daneshgah St, Tabriz, 51666-14766, Iran. .,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
105
|
Li N, Gao J, Mi L, Zhang G, Zhang L, Zhang N, Huo R, Hu J, Xu K. Synovial membrane mesenchymal stem cells: past life, current situation, and application in bone and joint diseases. Stem Cell Res Ther 2020; 11:381. [PMID: 32894205 PMCID: PMC7487958 DOI: 10.1186/s13287-020-01885-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 08/14/2020] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from not only bone marrow, but also various adult mesenchymal tissues such as periosteum, skeletal muscle, and adipose tissue. MSCs from different tissue sources have different molecular phenotypes and differentiation potential. Synovial membrane (SM) is an important and highly specific component of synovial joints. Previous studies have suggested that the synovium is a structure with a few cell layers thick and consists mainly of fibroblast-like synoviocytes (FLS), which forms a layer that lining the synovial membrane on the joint cavity and synovial fluid through cell-cell contact. In recent years, studies have found that there are also mesenchymal stem cells in the synovium, and as an important part of the mesenchymal stem cell family, it has strong capabilities of cartilage forming and tissue repairing. This article reviews the sources, surface markers, subtypes, influencing factors, and applications in inflammatory joints of synovial membrane mesenchymal stem cells (SM-MSCs) in recent years, aiming to clarify the research status and existing problems of SM-MSCs.
Collapse
Affiliation(s)
- Na Li
- Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Jinfang Gao
- Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Liangyu Mi
- Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Gailian Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Na Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Rongxiu Huo
- Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Junping Hu
- Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Ke Xu
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
106
|
Li Y, Yang J, Li M, Zhang X, Du J, Zhao X, Xu Z, Lin J. The Extracts of Human Fetal Brain Induce the Differentiation of Human Umbilical Cord Mesenchymal Stem Cells into Dopaminergic Neuron Containing Cells. Cell Reprogram 2020; 22:254-261. [PMID: 32833524 DOI: 10.1089/cell.2020.0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential to differentiate into neuron-like cells, which may provide a new strategy for the clinical treatment of neurodegenerative diseases such as Parkinson's disease (PD). However, the application of MSCs in the patients is still limited as the reason of efficiency and safety of transplantation. The aim of this study is to develop a new method and induce human umbilical cord MSCs (hUCMSCs) into neuron-like cells. Results from flow cytometry indicate that the isolated MSCs from hUCMSCs exhibited a typical phenotype of adult stem cells and express CD44, CD54, CD73, CD90, CD105, CD166, and HLA-ABC. Furthermore, the induced cells from hUCMSCs could spontaneously express different neural cell markers [neuron-specific enolase (NSE) and glial fibrillary acidic protein (GFAP)], even transcription factors related to dopaminergic neuron's development (Nurr1, Wnt-1, and En-1). Moreover, after treatment of EHFBT (extracts of human fetal brain tissue), hUCMSCs can express neuronal markers such as Nestin, LIM homeobox transcription factor 1 beta (LMX1B), dopamine beta hydroxylase (DBH), and dopamine transporter (DAT). In summary, a method that can induce hUCMSCs into dopaminergic neuron containing cells is established in vitro by the treatment of EHFBT. This would provide us a new cell source for PD in clinical treatment in the future.
Collapse
Affiliation(s)
- Yonghai Li
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Junzheng Yang
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Meng Li
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Xiaoyue Zhang
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jiang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xinghua Zhao
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Zhihao Xu
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
107
|
Liu J, Liu Q, Chen X. The Immunomodulatory Effects of Mesenchymal Stem Cells on Regulatory B Cells. Front Immunol 2020; 11:1843. [PMID: 32922398 PMCID: PMC7456948 DOI: 10.3389/fimmu.2020.01843] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022] Open
Abstract
The therapeutic potential of mesenchymal stem cells (MSCs) has been investigated in many preclinical and clinical studies. This potential is dominantly based on the immunosuppressive properties of MSCs. Although the therapeutic profiles of MSC transplantation are still not fully characterized, accumulating evidence has revealed that B cells change after MSC infusion, in particular inducing regulatory B cells (Bregs). The immunosuppressive effects of Bregs have been demonstrated, and these cells are being evaluated as new targets for the treatment of inflammatory diseases. MSCs are capable of educating B cells and inducing regulatory B cell production via cell-to-cell contact, soluble factors, and extracellular vesicles (EVs). These cells thus have the potential to complement each other's immunomodulatory functions, and a combined approach may enable synergistic effects for the treatment of immunological diseases. However, compared with investigations regarding other immune cells, investigations into how MSCs specifically regulate Bregs have been superficial and insufficient. In this review, we discuss the current findings related to the immunomodulatory effects of MSCs on regulatory B cells and provide optimal strategies for applications in immune-related disease treatments.
Collapse
Affiliation(s)
- Jialing Liu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiuli Liu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyong Chen
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
108
|
Yang JH, Liu FX, Wang JH, Cheng M, Wang SF, Xu DH. Mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles: Potential roles in rheumatic diseases. World J Stem Cells 2020; 12:688-705. [PMID: 32843922 PMCID: PMC7415241 DOI: 10.4252/wjsc.v12.i7.688] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been widely investigated in rheumatic disease due to their immunomodulatory and regenerative properties. Recently, mounting studies have implicated the therapeutic potency of MSCs mostly due to the bioactive factors they produce. Extracellular vesicles (EVs) derived from MSCs have been identified as a promising cell-free therapy due to low immunogenicity. Rheumatic disease, primarily including rheumatoid arthritis and osteoarthritis, is a group of diseases in which immune dysregulation and chronic progressive inflammation lead to irreversible joint damage. Targeting MSCs and MSC-derived EVs may be a more effective and promising therapeutic strategy for rheumatic diseases.
AIM To evaluate the potential therapeutic effectiveness of MSCs and EVs generated from MSCs in rheumatic diseases.
METHODS PubMed was searched for the relevant literature using corresponding search terms alone or in combination. Papers published in English language from January 1999 to February 2020 were considered. Preliminary screening of papers concerning analysis of "immunomodulatory function" or "regenerative function" by scrutinizing the titles and abstracts of the literature, excluded the papers not related to the subject of the article. Some other related studies were obtained by manually retrieving the reference lists of papers that comply with the selection criteria, and these studies were screened to meet the final selection and exclusion criteria.
RESULTS Eighty-six papers were ultimately selected for analysis. After analysis of the literature, it was found that both MSCs and EVs generated from MSCs have great potential in multiple rheumatic diseases, such as rheumatoid arthritis and osteoarthritis, in repair and regeneration of tissues, inhibition of inflammatory response, and regulation of body immunity via promoting chondrogenesis, regulating innate and adaptive immune cells, and regulating the secretion of inflammatory factors. But EVs from MSCs exhibit much more advantages over MSCs, which may represent another promising cell-free restorative strategy. Targeting MSCs and MSC-derived EVs may be a more efficient treatment for patients with rheumatic diseases.
CONCLUSION The enormous potential of MSCs and EVs from MSCs in immunomodulation and tissue regeneration offers a new idea for the treatment of rheumatism. However, more in-depth exploration is needed before their clinical application.
Collapse
Affiliation(s)
- Jing-Han Yang
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Feng-Xia Liu
- Department of Allergy, Weifang People’s Hospital, Weifang 261000, Shandong Province, China
| | - Jing-Hua Wang
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Min Cheng
- Department of Physiology, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Shu-Feng Wang
- Medical Experimental Training Center, Weifang Medical University, Weifang 261000, Shandong Province, China
| | - Dong-Hua Xu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
- Department of Rheumatology of the First Affiliated Hospital, Weifang Medical University, Weifang 261000, Shandong Province, China
| |
Collapse
|
109
|
The Effects of Mesenchymal Stem Cells on Antimelanoma Immunity Depend on the Timing of Their Administration. Stem Cells Int 2020; 2020:8842659. [PMID: 32695181 PMCID: PMC7368936 DOI: 10.1155/2020/8842659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is still a lively debate about whether mesenchymal stem cells (MSCs) promote or suppress antitumor immune response. Although several possible explanations have been proposed, including different numbers of injected and engrafted MSCs, heterogeneity in phenotype, and function of tumor cells, the exact molecular mechanisms responsible for opposite effects of MSCs in modulation of antitumor immunity are still unknown. Herewith, we used a B16F10 murine melanoma model to investigate whether timing of MSC administration in tumor-bearing mice was crucially important for their effects on antitumor immunity. MSCs, intravenously injected 24 h after melanoma induction (B16F10+MSC1d-treated mice), significantly enhanced natural killer (NK) and T cell-driven antitumor immunity, suppressed tumor growth, and improved survival of melanoma-bearing animals. Significantly higher plasma levels of antitumorigenic cytokines (TNF-α and IFN-γ), remarkably lower plasma levels of immunosuppressive cytokines (TGF-β and IL-10), and a significantly higher number of tumor-infiltrating, IFN-γ-producing, FasL- and granzyme B-expressing NK cells, IL-17-producing CD4+Th17 cells, IFN-γ- and TNF-α-producing CD4+Th1 cells, and CD8+cytotoxic T lymphocytes (CTLs) were observed in B16F10+MSC1d-treated mice. On the contrary, MSCs, injected 14 days after melanoma induction (B16F10+MSC14d-treated mice), promoted tumor growth by suppressing antigen-presenting properties of tumor-infiltrating dendritic cells (DCs) and macrophages and by reducing tumoricidal capacity of NK cells and T lymphocytes. Significantly higher plasma levels of TGF-β and IL-10, remarkably lower plasma levels of TNF-α and IFN-γ, and significantly reduced number of tumor-infiltrating, I-A-expressing, and IL-12-producing macrophages, CD80- and I-A-expressing DCs, granzyme B-expressing CTLs and NK cells, IFN-γ- and IL-17-producing CTLs, CD4+Th1, and Th17 cells were observed in B16F10+MSC14d-treated animals. In summing up, the timing of MSC administration into the tumor microenvironment was crucially important for MSC-dependent modulation of antimelanoma immunity. MSCs transplanted during the initial phase of melanoma growth exerted tumor-suppressive effect, while MSCs injected during the progressive stage of melanoma development suppressed antitumor immunity and enhanced tumor expansion.
Collapse
|
110
|
Nie H, An F, Mei J, Yang C, Zhan Q, Zhang Q. IL-1 β Pretreatment Improves the Efficacy of Mesenchymal Stem Cells on Acute Liver Failure by Enhancing CXCR4 Expression. Stem Cells Int 2020; 2020:1498315. [PMID: 32724311 PMCID: PMC7364198 DOI: 10.1155/2020/1498315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/15/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), with the powerful metabolic and functional supporting abilities for inflammatory diseases, may be an effective therapeutic strategy for acute liver failure (ALF). However, the efficacy of MSCs can still be promoted if pretreatment is applied to enhance their poor migration towards the damaged liver. The purpose of this study is to determine the effect of IL-1β pretreatment on the efficacy and homing ability of MSCs in ALF. METHODS MSCs were isolated by the whole bone marrow adherence method and characterized. The efficacy and homing ability of IL-1β-pretreated MSCs (Pre-MSCs) were examined in a rat ALF model and compared with that of MSCs and normal saline. Then, Western blot was performed to detect the c-Met and CXCR4 expression of MSCs and Pre-MSCs and followed by flow cytometry to detect the meaningful indicators. Finally, the migration abilities of different cells and different conditions were tested by the Transwell migration assay. RESULTS MSCs of ideal purity were successfully isolated and cultured. Comparing with MSCs, Pre-MSCs had significantly better efficacy on improving the survival rate and liver function of ALF rats. Further analyses of damaged liver tissues showed that IL-1β pretreatment significantly enhanced the efficacy of MSCs on suppressing liver necrosis. Besides, Pre-MSCs exhibited better effects in inhibiting apoptosis and activating proliferation. The results of tracing experiments with CM-Dil-labeled cells confirmed that more cells migrated to the damaged liver in the Pre-MSC group. In terms of mechanism, the CXCR4 expression was significantly enhanced by IL-1β pretreatment, and an increased migration ability towards SDF-1 that could be reversed by AMD3100 was found in Pre-MSCs. CONCLUSION IL-1β pretreatment could enhance the homing ability of MSCs at least partially by increasing the expression of CXCR4 and further improve the efficacy of MSCs on ALF.
Collapse
Affiliation(s)
- He Nie
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Fangmei An
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Cheng Yang
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Qiang Zhan
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Qinglin Zhang
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| |
Collapse
|
111
|
Cao J, Wang B, Tang T, Lv L, Ding Z, Li Z, Hu R, Wei Q, Shen A, Fu Y, Liu B. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res Ther 2020; 11:206. [PMID: 32460853 PMCID: PMC7251891 DOI: 10.1186/s13287-020-01719-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/16/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023] Open
Abstract
Background Exosomes derived from mesenchymal stem cells (MSC-exos) have been demonstrated with great potential in the treatment of multiple human diseases including acute kidney injury (AKI) by virtue of their intrinsic cargoes. However, there are major challenges of low yield and the lack of an established biomanufacturing platform to efficiently produce MSC-exos, thereby limiting their therapeutic application. Here, we aimed to establish a novel strategy to produce MSC-exos with a hollow fiber bioreactor-based three-dimensional (3D) culture system and evaluate the therapeutic efficacy of 3D-exosomes (3D-exos) on AKI. Methods Mesenchymal stem cells (MSCs) were isolated from fresh human umbilical cord and cultured in two-dimensional (2D) flasks. 2 × 108 MSCs were inoculated into the hollow fiber bioreactor for 3D culture. The culture supernatants were collected every 1 or 2 days for isolating exosomes. Exosomes from 2D (2D-exos) and 3D cultures were characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting analysis of exosome markers. The yield of exosomes from 2 × 108 MSCs seeded in 2D and 3D culture system was compared, based on protein quantification. The therapeutic efficacy of 2D-exos and 3D-exos was investigated in a murine model of cisplatin-induced AKI in vivo and in vitro. Results 3D culture did not significantly change the surface markers of MSCs, as well as the morphology, size, and exosomal markers of 3D-exos when compared to those of 2D-exos. Compared with conventional 2D culture, the 3D culture system increased total exosome production up to 19.4-fold. 3D-exos were more concentrated in the harvested supernatants (15.5-fold) than 2D-exos, which led to a higher exosome collection efficiency of 3D culture system. In vivo, both 2D-exos and 3D-exos significantly alleviated cisplatin-induced murine AKI evidenced by improved renal function, attenuated pathological changes of renal tubules, reduced inflammatory factors, and repressed T cell and macrophage infiltration. Impressively, 3D-exos were more effective than 2D-exos. Moreover, 3D-exos were taken up by tubular epithelial cells (TECs) with improved efficiency, thereby exhibiting superior anti-inflammatory effect and improved viability of TECs in vitro. Conclusions In summary, our findings demonstrate that the hollow fiber 3D culture system provides an efficient strategy for the continuous production of MSC-exos which has enhanced therapeutic potential for cisplatin-induced AKI.
Collapse
Affiliation(s)
- Jingyuan Cao
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Taotao Tang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Linli Lv
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Zhaoying Ding
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Zuolin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Ruoyu Hu
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Qing Wei
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Anran Shen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Yuqi Fu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
112
|
Autologous Mesenchymal Stem Cells Improve Motor Recovery in Subacute Ischemic Stroke: a Randomized Clinical Trial. Transl Stroke Res 2020; 11:910-923. [PMID: 32462427 DOI: 10.1007/s12975-020-00787-z] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022]
Abstract
While preclinical stroke studies have shown that mesenchymal stem cells (MSCs) promote recovery, few randomized controlled trials (RCT) have assessed cell therapy in humans. In this RCT, we assessed the safety, feasibility, and efficacy of intravenous autologous bone marrow-derived MSCs in subacute stroke. ISIS-HERMES was a single-center, open-label RCT, with a 2-year follow-up. We enrolled patients aged 18-70 years less than 2 weeks following moderate-severe ischemic carotid stroke. Patients were randomized 2:1 to receive intravenous MSCs or not. Primary outcomes assessed feasibility and safety. Secondary outcomes assessed global and motor recovery. Passive wrist movement functional MRI (fMRI) activity in primary motor cortex (MI) was employed as a motor recovery biomarker. We compared "treated" and "control" groups using as-treated analyses. Of 31 enrolled patients, 16 patients received MSCs. Treatment feasibility was 80%, and there were 10 and 16 adverse events in treated patients, and 12 and 24 in controls at 6-month and 2-year follow-up, respectively. Using mixed modeling analyses, we observed no treatment effects on the Barthel Index, NIHSS, and modified-Rankin scores, but significant improvements in motor-NIHSS (p = 0.004), motor-Fugl-Meyer scores (p = 0.028), and task-related fMRI activity in MI-4a (p = 0.031) and MI-4p (p = 0.002). Intravenous autologous MSC treatment following stroke was safe and feasible. Motor performance and task-related MI activity results suggest that MSCs improve motor recovery through sensorimotor neuroplasticity. ClinicalTrials.gov Identifier NCT00875654.
Collapse
|
113
|
Harrell CR, Jovicic N, Djonov V, Volarevic V. Therapeutic Use of Mesenchymal Stem Cell-Derived Exosomes: From Basic Science to Clinics. Pharmaceutics 2020; 12:pharmaceutics12050474. [PMID: 32456070 PMCID: PMC7313713 DOI: 10.3390/pharmaceutics12050474] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSC) are, due to their immunosuppressive and regenerative properties, used as new therapeutic agents in cell-based therapy of inflammatory and degenerative diseases. A large number of experimental and clinical studies revealed that most of MSC-mediated beneficial effects were attributed to the effects of MSC-sourced exosomes (MSC-Exos). MSC-Exos are nano-sized extracellular vesicles that contain MSC-derived bioactive molecules (messenger RNA (mRNA), microRNAs (miRNAs)), enzymes, cytokines, chemokines, and growth factors) that modulate phenotype, function and homing of immune cells, and regulate survival and proliferation of parenchymal cells. In this review article, we emphasized current knowledge about molecular and cellular mechanisms that were responsible for MSC-Exos-based beneficial effects in experimental models and clinical trials. Additionally, we elaborated on the challenges of conventional MSC-Exos administration and proposed the use of new bioengineering and cellular modification techniques which could enhance therapeutic effects of MSC-Exos in alleviation of inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Carl Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, FL 34684, USA;
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia;
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, 2 Baltzerstrasse, 3012 Bern, Switzerland;
| | - Vladislav Volarevic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), Università di Catania, Via Santa Sofia 78, 95123 Catania, Italy
- Correspondence: ; Tel.: +381-34306800; Fax: +381-34306800
| |
Collapse
|
114
|
Sveiven SN, Nordgren TM. Lung-resident mesenchymal stromal cells are tissue-specific regulators of lung homeostasis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L197-L210. [PMID: 32401672 DOI: 10.1152/ajplung.00049.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Until recently, data supporting the tissue-resident status of mesenchymal stromal cells (MSC) has been ambiguous since their discovery in the 1950-60s. These progenitor cells were first discovered as bone marrow-derived adult multipotent cells and believed to migrate to sites of injury, opposing the notion that they are residents of all tissue types. In recent years, however, it has been demonstrated that MSC can be found in all tissues and MSC from different tissues represent distinct populations with differential protein expression unique to each tissue type. Importantly, these cells are efficient mediators of tissue repair, regeneration, and prove to be targets for therapeutics, demonstrated by clinical trials (phase 1-4) for MSC-derived therapies for diseases like graft-versus-host-disease, multiple sclerosis, rheumatoid arthritis, and Crohn's disease. The tissue-resident status of MSC found in the lung is a key feature of their importance in the context of disease and injuries of the respiratory system, since these cells could be instrumental to providing more specific and targeted therapies. Currently, bone marrow-derived MSC have been established in the treatment of disease, including diseases of the lung. However, with lung-resident MSC representing a unique population with a different phenotypic and gene expression pattern than MSC derived from other tissues, their role in remediating lung inflammation and injury could provide enhanced efficacy over bone marrow-derived MSC methods. Through this review, lung-resident MSC will be characterized, using previously published data, by surface markers, gene expression patterns, and compared with bone-marrow MSC to highlight similarities and, importantly, differences in these cell types.
Collapse
Affiliation(s)
- Stefanie Noel Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| |
Collapse
|
115
|
Nano-Ghosts: Biomimetic membranal vesicles, technology and characterization. Methods 2020; 177:126-134. [DOI: 10.1016/j.ymeth.2019.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022] Open
|
116
|
Atkinson SP. A Preview of Selected Articles. Stem Cells 2020; 38:587-589. [PMID: 32333501 DOI: 10.1002/stem.3184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 11/08/2022]
|
117
|
Zhang F, Wang C, Wen X, Chen Y, Mao R, Cui D, Li L, Liu J, Chen Y, Cheng J, Lu Y. Mesenchymal stem cells alleviate rat diabetic nephropathy by suppressing CD103 + DCs-mediated CD8 + T cell responses. J Cell Mol Med 2020; 24:5817-5831. [PMID: 32283569 PMCID: PMC7214166 DOI: 10.1111/jcmm.15250] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/27/2020] [Accepted: 03/15/2020] [Indexed: 02/05/2023] Open
Abstract
Diabetic nephropathy (DN) as a kind of serious microvascular complication of Diabetes Mellitus (DM) usually causes the end‐stage of renal disease (ESRD). Studies have demonstrated that CD103+ dendritic cells (DCs) exhibited a renal pathogenic effect in murine chronic kidney disease (CKD). Mesenchymal stem cells (MSCs) can alleviate DN and suppress the DCs maturation. To explore the role of CD103+ DCs and the potential mechanisms underlying MSCs‐mediated protective effects in DN, we used bone marrow MSCs (BM‐MSCs) to treat DN rats. MSCs transplantation considerably recovered kidney function and diminished renal injury, fibrosis and the population of renal CD103+ DCs in DN rat. The MSCs‐treated DN rats had decreased mRNA expression levels of interleukin (IL)1β, IL6, tumour necrosis factor alpha (TNF‐α), monocyte chemotactic protein 1 (MCP‐1) and reduced CD8 T cell infiltration in the kidney. MSCs significantly down‐regulated the genes expression of transcription factors (Basic leucine zipper transcriptional factor ATF‐like 3, Batf3 and DNA‐binding protein inhibitor ID‐2, Id2) and FMS‐like tyrosine kinase‐3 (Flt3) which are necessary for CD103+ DCs development. The protective effect of MSCs may be partly related to their immunosuppression of CD8+ T cell proliferation and activation mediated by CD103+ DCs in the kidney of DN rats.
Collapse
Affiliation(s)
- Fuping Zhang
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chengshi Wang
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wen
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Chen
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiwen Mao
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Danli Cui
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
118
|
Cao Y, Ji C, Lu L. Mesenchymal stem cell therapy for liver fibrosis/cirrhosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:562. [PMID: 32775363 PMCID: PMC7347778 DOI: 10.21037/atm.2020.02.119] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Liver fibrosis represents a common outcome of most chronic liver diseases. Advanced fibrosis leads to cirrhosis for which no effective treatment is available except liver transplantation. Because of the limitations of liver transplantation, alternative therapeutic strategies are an urgent need to find. Recently, mesenchymal stem cells (MSCs) based therapy has been suggested as an attractive therapeutic option for liver fibrosis and cirrhosis, based on the promising results from preclinical and clinical studies. Although the precise mechanisms of MSC transplantation are still not fully understood, accumulating evidence has indicated that MSCs eliminate the progression of fibrosis due to their immune-modulatory properties. In this review, we summarise the properties of MSCs and their clinical application in the treatment of liver fibrosis and cirrhosis. We also discuss the mechanisms involved in MSC-dependent regulation of immune microenvironment in the context of liver fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Yan Cao
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Ling Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing 210029, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
119
|
Fan Y, Herr F, Vernochet A, Mennesson B, Oberlin E, Durrbach A. Human Fetal Liver Mesenchymal Stem Cell-Derived Exosomes Impair Natural Killer Cell Function. Stem Cells Dev 2020; 28:44-55. [PMID: 30328799 DOI: 10.1089/scd.2018.0015] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are powerful immunomodulators that regulate the diverse functions of immune cells involved in allogeneic reactions, such as T cells and natural killer (NK) cells, through cell-cell contact or secreted factors. Exosomes secreted by MSCs may be involved in their regulatory functions, providing new therapeutic tools. Here, we showed that fetal liver (FL) MSC-derived exosomes inhibit proliferation, activation, and cytotoxicity of NK cells. Exosomes bearing latency associated peptide (LAP), TGFβ, and thrombospondin 1 (TSP1), a regulatory molecule for TGFβ, induced downstream TGFβ/Smad2/3 signaling in NK cells. The inhibition of TGFβ, using a neutralizing anti-TGFβ antibody, restored NK proliferation, differentiation, and cytotoxicity, demonstrating that FL-MSC-derived exosomes exert their inhibition on NK cell function via TGFβ. These results suggest that FL-MSC-derived exosomes regulate NK cell functions through exosome-associated TGFβ.
Collapse
Affiliation(s)
- Ye Fan
- 1 INSERM UMR-S1197, Villejuif, France
| | | | | | - Benoît Mennesson
- 2 Service de Gynécologie-Obstétrique, Hôpital René-Dubos, Pontoise, France
| | | | - Antoine Durrbach
- 1 INSERM UMR-S1197, Villejuif, France
- 3 Département de Néphrologie, Hôpital Le Kremlin Bicêtre, IFRNT, Université Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
120
|
Harrell CR, Markovic BS, Fellabaum C, Arsenijevic N, Djonov V, Volarevic V. The role of Interleukin 1 receptor antagonist in mesenchymal stem cell-based tissue repair and regeneration. Biofactors 2020; 46:263-275. [PMID: 31755595 DOI: 10.1002/biof.1587] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/27/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022]
Abstract
Interleukin (IL)-1 receptor antagonist (IL-1Ra), a naturally occurring antagonist of IL-1α/IL-1β signaling pathways, has been attributed to the immunosuppressive effects of mesenchymal stem cells (MSCs). MSCs, in IL-1Ra-dependent manner, suppressed production of IL-1β in dermal macrophages, induced their polarization in anti-inflammatory M2 phenotype, attenuated antigen-presenting properties of dendritic cells (DCs), and promoted expansion of immunosuppressive T regulatory cells in the skin, which resulted in enhanced repair of the nonhealing wounds. Reduced activation of inflammasome and suppressed production of IL-1β in macrophages were mainly responsible for beneficial effects of MSC-derived IL-1Ra in alleviation of acute lung injury, dry eye syndrome, and corneal injury. Through the production of IL-1Ra, MSCs reduced migration of DCs to the draining lymph nodes and attenuated generation of inflammatory Th1 and Th17 cells that resulted in alleviation of fulminant hepatitis and rheumatoid arthritis. MSCs, in IL-1Ra-dependent manner, reduced liver fibrosis by suppressing production of Type I collagen in hepatic stellate cells. IL-1Ra was, at least partially, responsible for enhanced proliferation of hepatocytes and chondrocytes in MSC-treated animals with partial hepatectomy and osteoarthritis. Despite of these beneficial effects, IL-1Ra-dependent inhibition of IL-1α/IL-1β-signaling significantly increased risk of infections. Therefore, future experimental and clinical studies should delineate potential side effects of MSC-derived IL-1Ra before IL-1Ra-overexpressing MSCs could be used as a potentially new therapeutic agent for the treatment of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Bojana Simovic Markovic
- Faculty of Medical Sciences, Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | | | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | | | - Vladislav Volarevic
- Faculty of Medical Sciences, Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
121
|
Messina A, Luce E, Hussein M, Dubart-Kupperschmitt A. Pluripotent-Stem-Cell-Derived Hepatic Cells: Hepatocytes and Organoids for Liver Therapy and Regeneration. Cells 2020; 9:cells9020420. [PMID: 32059501 PMCID: PMC7072243 DOI: 10.3390/cells9020420] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
The liver is a very complex organ that ensures numerous functions; it is thus susceptible to multiple types of damage and dysfunction. Since 1983, orthotopic liver transplantation (OLT) has been considered the only medical solution available to patients when most of their liver function is lost. Unfortunately, the number of patients waiting for OLT is worryingly increasing, and extracorporeal liver support devices are not yet able to counteract the problem. In this review, the current and expected methodologies in liver regeneration are briefly analyzed. In particular, human pluripotent stem cells (hPSCs) as a source of hepatic cells for liver therapy and regeneration are discussed. Principles of hPSC differentiation into hepatocytes are explored, along with the current limitations that have led to the development of 3D culture systems and organoid production. Expected applications of these organoids are discussed with particular attention paid to bio artificial liver (BAL) devices and liver bio-fabrication.
Collapse
Affiliation(s)
- Antonietta Messina
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Eléanor Luce
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Marwa Hussein
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
- Correspondence: ; Tel.: +33-145595138
| |
Collapse
|
122
|
Hu C, Wu Z, Li L. Mesenchymal stromal cells promote liver regeneration through regulation of immune cells. Int J Biol Sci 2020; 16:893-903. [PMID: 32071558 PMCID: PMC7019139 DOI: 10.7150/ijbs.39725] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is sensitive to pathogen-induced acute or chronic liver injury, and liver transplantation (LT) is the only effective strategy for end-stage liver diseases. However, the clinical application is limited by a shortage of liver organs, immunological rejection and high cost. Mesenchymal stromal cell (MSC)-based therapy has gradually become a hot topic for promoting liver regeneration and repairing liver injury in various liver diseases, since MSCs are reported to migrate toward injured tissues, undergo hepatogenic differentiation, inhibit inflammatory factor release and enhance the proliferation of liver cells in vivo. MSCs exert immunoregulatory effects through cell-cell contact and the secretion of anti-inflammatory factors to inhibit liver inflammation and promote liver regeneration. In addition, MSCs are reported to effectively inhibit the activation of cells of the innate immune system, including macrophages, natural killer (NK) cells, dendritic cells (DCs), monocytes and other immune cells, and inhibit the activation of cells of the adaptive immune system, including T lymphocytes, B lymphocytes and subsets of T cells or B cells. In the current review, we mainly focus on the potential effects and mechanisms of MSCs in inhibiting the activation of immune cells to attenuate liver injury in models or patients with acute liver failure (ALF), nonalcoholic fatty liver disease (NAFLD), and liver fibrosis and in patients or models after LT. We highlight that MSC transplantation may replace general therapies for eliminating acute or chronic liver injury in the near future.
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, PR China
| | - Zhongwen Wu
- 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, PR 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, PR China
| |
Collapse
|
123
|
Gonzalez-Pujana A, Igartua M, Santos-Vizcaino E, Hernandez RM. Mesenchymal stromal cell based therapies for the treatment of immune disorders: recent milestones and future challenges. Expert Opin Drug Deliv 2020; 17:189-200. [PMID: 31918562 DOI: 10.1080/17425247.2020.1714587] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Mesenchymal stromal cells (MSCs) present unique immunomodulatory properties that make them promising candidates for the treatment of inflammatory and immune disorders. MSC-mediated immunomodulation is a complex combination of mechanisms, in which the secretome plays a fundamental role. The plethora of bioactive molecules MSCs produce, such as indoleamine 2,3-dioxygenase (IDO) or prostaglandin E2 (PGE2), efficiently regulates innate and adaptive immunity. As a result, MSCs have been extensively employed in preclinical studies, leading to the conduction of multiple clinical trials.Areas covered: This review summarizes the effects of some of the key biomolecules in the MSC secretome and the advances in preclinical studies exploring the treatment of disorders including graft-versus-host disease (GvHD) or inflammatory bowel disease (IBD). Further, late-stage clinical trials and the first MSC-based therapies that recently obtained regulatory approval are discussed.Expert opinion: Extensive research supports the potential of MSC-based immunomodulatory therapies. However, to establish the bases for clinical translation, the future of study lies in the standardization of protocols and in the development of strategies that boost the therapeutic properties of MSCs.
Collapse
Affiliation(s)
- Ainhoa Gonzalez-Pujana
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country, UPV/EHU, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country, UPV/EHU, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country, UPV/EHU, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country, UPV/EHU, Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| |
Collapse
|
124
|
Xiang YJ, Hou YY, Yan HL, Liu H, Ge YX, Chen N, Xiang JF, Hao CF. Mesenchymal stem cells-derived exosomes improve pregnancy outcome through inducing maternal tolerance to the allogeneic fetus in abortion-prone mating mouse. Kaohsiung J Med Sci 2020; 36:363-370. [PMID: 31943723 DOI: 10.1002/kjm2.12178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is three or more times of consecutive spontaneous loss of pregnancy. The underlying cause is complicated and the etiology of over 50% of RPL patients is unclear. In the present study, bone marrow mesenchymal stem cells were isolated from CBA/J female mice and exosomes were isolated from cell culture medium by ultracentrifugation. CBA/J female mice were paired with male DBA/2 to generate abortion prone mouse model, and CBA/J females paired with male BALB/c mice were used as control. Exosomes were injected through uterine horns into pregnant CBA/J mice on day 4.5 of gestation in abortion-prone matting. On day 13.5 of pregnancy, abortion rates were calculated and the level of transforming growth factor-β (TGF-β), interleukin 10 (IL-10), interferon g (IFN-γ), and tumor necrosis factor a (TNF-α) in CD4+ T cells and macrophages in deciduas were evaluated by flow cytometry. Exosomes injection improved the pregnancy outcomes in abortion prone mice. The IL-4 and IL-10 levels on CD4+ T cells were upregulated in the maternal-fetal interface; meanwhile, the TNF-α and IFN-γ levels on CD4+ T cells were reduced. The IL-10 level was increased and IL-12 was reduced on the monocytes that separated from deciduas. miR-101 level was increased in the CD4+ T cells in the deciduas. In conclusion, the treatment of ESCs-derived exosomes modulates T cells' function and macrophages activities in the maternal-fetal interface that resulted in a decreased embryo resorption rate, and provides a therapeutic potential to treat RPL.
Collapse
Affiliation(s)
- Yan-Jie Xiang
- School of Medicine, Shandong University, Jinan, Shandong Province, People's Republic of China.,Reproductive Medicine Center, The Affiliated Changhai Hospital of China Naval Military Medical University, Shanghai, People's Republic of China
| | - Yan-Yan Hou
- Maternity Department of International Maternal and Child Peace Hospital, The Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hong-Li Yan
- Reproductive Medicine Center, The Affiliated Changhai Hospital of China Naval Military Medical University, Shanghai, People's Republic of China
| | - Hui Liu
- Department of Reproductive Medicine, People's Hospital of Rizhao, Rizhao, Shandong, People's Republic of China
| | - Yan-Xin Ge
- Department of Reproductive Medicine, People's Hospital of Rizhao, Rizhao, Shandong, People's Republic of China
| | - Na Chen
- Department of Obstetrics, Traditional Chinese Medicine Hospital of The West Coast of Qingdao New District, Qingdao, Shandong, People's Republic of China
| | - Jian-Feng Xiang
- Department of Intervention, Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China
| | - Cui-Fang Hao
- Center for Reproductive Medicine, Qingdao Women and Children's Hospital (QWCH) affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
125
|
Yuan Y, Li L, Zhu L, Liu F, Tang X, Liao G, Liu J, Cheng J, Chen Y, Lu Y. Mesenchymal stem cells elicit macrophages into M2 phenotype via improving transcription factor EB-mediated autophagy to alleviate diabetic nephropathy. Stem Cells 2020; 38:639-652. [PMID: 31904160 DOI: 10.1002/stem.3144] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023]
Abstract
Diabetic nephropathy (DN) is a leading cause of end-stage renal disease. Chronic inflammation is recognized as a key causal factor in the development and progression of DN, and the imbalance of M1/M2 macrophages (Mφ) contributes to this process. Mesenchymal stem cells (MSCs) have been reported to prevent renal injuries via immune regulation in diabetic models, but whether these benefits are owing to the regulation of Mφ, and the underlying signaling pathways are unknown. Here, we showed that MSCs elicited Mφ into M2 phenotype and prevented renal injuries in DN mice, but these effects were abolished when the Mφ were depleted by clodronate liposomes (Lipo-Clod), suggesting that Mφ were necessary for renal protection of MSCs in DN mice. Moreover, the MSCs promoted M2 polarization was attributable to the activation of transcription factor EB (TFEB) and subsequent restore of lysosomal function and autophagy activity in Mφ. Furthermore, in vivo adoptive transfer of Mφin vivo (Mφ from DN + MSCs mice) or MφMSCs (Mφ cocultured with MSCs in vitro) to DN mice improved renal function. While, TFEB knockdown in Mφ significantly abolished the protective role of MφMSCs . Altogether, these findings revealed that MSCs suppress inflammatory response and alleviate renal injuries in DN mice via TFEB-dependent Mφ switch.
Collapse
Affiliation(s)
- Yujia Yuan
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lan Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lingling Zhu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Fei Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xi Tang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Guangneng Liao
- Animal Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingqiu Cheng
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Younan Chen
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yanrong Lu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
126
|
Therapeutic Potential of Mesenchymal Stem Cells and Their Secretome in the Treatment of Glaucoma. Stem Cells Int 2019; 2019:7869130. [PMID: 31949441 PMCID: PMC6948292 DOI: 10.1155/2019/7869130] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Glaucoma represents a group of progressive optic neuropathies characterized by gradual loss of retinal ganglion cells (RGCs), the neurons that conduct visual information from the retina to the brain. Elevated intraocular pressure (IOP) is considered the main reason for enhanced apoptosis of RGCs in glaucoma. Currently used therapeutic agents are not able to repopulate and/or regenerate injured RGCs and, therefore, are ineffective in most patients with advanced glaucoma. Accordingly, several new therapeutic approaches, including stem cell-based therapy, have been explored for the glaucoma treatment. In this review article, we emphasized current knowledge regarding molecular and cellular mechanisms responsible for beneficial effects of mesenchymal stem cells (MSCs) and their secretome in the treatment of glaucoma. MSCs produce neurotrophins and in an exosome-dependent manner supply injured RGCs with growth factors enhancing their survival and regeneration. Additionally, MSCs are able to generate functional RGC-like cells and induce proliferation of retinal stem cells. By supporting integrity of trabecular meshwork, transplanted MSCs alleviate IOP resulting in reduced loss of RGCs. Moreover, MSCs are able to attenuate T cell-driven retinal inflammation providing protection to the injured retinal tissue. In summing up, due to their capacity for neuroprotection and immunomodulation, MSCs and their secretome could be explored in upcoming clinical studies as new therapeutic agents for glaucoma treatment.
Collapse
|
127
|
Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells 2019; 8:cells8121605. [PMID: 31835680 PMCID: PMC6952783 DOI: 10.3390/cells8121605] [Citation(s) in RCA: 512] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/24/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
Abstract
There is growing evidence that mesenchymal stem cell (MSC)-based immunosuppression was mainly attributed to the effects of MSC-derived extracellular vesicles (MSC-EVs). MSC-EVs are enriched with MSC-sourced bioactive molecules (messenger RNA (mRNA), microRNAs (miRNAs), cytokines, chemokines, immunomodulatory factors) that regulate phenotype, function and homing of immune cells. In this review article we emphasized current knowledge regarding molecular mechanisms responsible for the therapeutic effects of MSC-EVs in attenuation of autoimmune and inflammatory diseases. We described the disease-specific cellular targets of MSC-EVs and defined MSC-sourced molecules, which were responsible for MSC-EV-based immunosuppression. Results obtained in a large number of experimental studies revealed that both local and systemic administration of MSC-EVs efficiently suppressed detrimental immune response in inflamed tissues and promoted survival and regeneration of injured parenchymal cells. MSC-EVs-based anti-inflammatory effects were relied on the delivery of immunoregulatory miRNAs and immunomodulatory proteins in inflammatory immune cells (M1 macrophages, dendritic cells (DCs), CD4+Th1 and Th17 cells), enabling their phenotypic conversion into immunosuppressive M2 macrophages, tolerogenic DCs and T regulatory cells. Additionally, through the delivery of mRNAs and miRNAs, MSC-EVs activated autophagy and/or inhibited apoptosis, necrosis and oxidative stress in injured hepatocytes, neurons, retinal cells, lung, gut and renal epithelial cells, promoting their survival and regeneration.
Collapse
|
128
|
Abo-Aziza FA, Zaki AKA, Abo El-Maaty AM. Bone marrow-derived mesenchymal stem cell (BM-MSC): A tool of cell therapy in hydatid experimentally infected rats. CELL REGENERATION (LONDON, ENGLAND) 2019; 8:58-71. [PMID: 31844519 PMCID: PMC6895685 DOI: 10.1016/j.cr.2019.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022]
Abstract
This study aimed to clarify the potentiality of bone marrow mesenchymal stem cells (BM-MSC) transplantation with albendazole (ABZ) on the modulation of immune responses against hydatid cyst antigens and the regeneration of injured livers in experimentally infected rats. Three different antigens of hydatid cyst fluid (HCF), hydatid cyst protoscolex (HCP) and hydatid cyst germinal layer (HCG) were isolated and their antigenic potencies were determined. The ultrasound, immunological and pathological criteria were investigated. Counting of 80% confluence BM-MSC was 4.68 × 104 cells/cm2 with 92.24% viability. Final population doublings score was 65.31 that indicated proliferation and self-renewability. Phenotyping of BM-MSC showed expression of CD73 and CD29 without exhibition of CD34 and CD14. Ultrasound examination showed multiple hydatid cysts in liver with low blood flow and spleenomegaly 8 weeks' post infection. No significant differences were noted in cystic diameter in uni-cyst liver at 2nd and 4th weeks following ABZ treatment while it was significantly decreased (P < 0.05) following transplantation of BM-MSC + ABZ treatment comparing to experimentally infected untreated group. Igs and IgG responses to the three antigens were significantly elevated while elevation in IgM response was only to HCG (P < 0.05). ABZ treatment accompanied with significant decrease in Igs and IgG titers against HCF and HCG only at 4th week post treatment (P < 0.05). However, Igs titer against HCF, HCP and HCG was significantly decreased at the 4th week following transplantation of BM-MSC + ABZ. Interestingly, the combination of BM-MSC + ABZ treatment resulted in reduction of Igs response to HCP to normal level as that of healthy control. Experimental infection resulted in elevation of TNF-α and IL-6 (P < 0.05) while, IL-4 and IL-10 decreased (P < 0.01). After transplantation of BM-MSC + ABZ treatment, serum TNF-α and IL-6 concentrations were reduced (P < 0.05) at both the 2nd and 4th weeks. However, IL-4 and IL-10 concentrations were significantly elevated (P < 0.05) only at 4th week following transplantation of BM-MSC + ABZ treatment. In conclusion, BM-MSC transplantation following ABZ administration can regenerate injured liver tissue without complete disappearance of hydatid cyst. In addition, it can modulate host protective humeral and cell mediated immune responses against hydatid cyst antigens. Therefore, the current study encourages to move to the step of performing clinical trials in humans.
Collapse
Affiliation(s)
- Faten A.M. Abo-Aziza
- Department of Parasitology and Animal Diseases, Veterinary Research Division, National Research Centre, Cairo, Egypt
| | - Abdel Kader A. Zaki
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Amal M. Abo El-Maaty
- Department of Animal Reproduction and AI, Veterinary Research Division, National Research Centre, Cairo, Egypt
| |
Collapse
|
129
|
Therapeutic Potential of “Exosomes Derived Multiple Allogeneic Proteins Paracrine Signaling: Exosomes d-MAPPS” is Based on the Effects of Exosomes, Immunosuppressive and Trophic Factors. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2018-0032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Due to their differentiation capacity and potent immunosuppressive and pro-angiogenic properties, mesenchymal stem cells (MSCs) have been considered as new therapeutic agents in regenerative medicine. Since most of MSC-mediated beneficent effects are a consequence of their paracrine action, we designed MSC-based product “Exosomes Derived Multiple Allogeneic Proteins Paracrine Signaling (Exosomes d-MAPPS), which activity is based on MSCs-derived growth factors and immunomodulatory cytokines capable to attenuate inflammation and to promote regeneration of injured tissues. Interleukin 1 receptor antagonist (IL-1Ra) and IL-27 were found in high concentrations in Exosomes d-MAPPS samples indicating strong anti-inflammatory and immunosuppressive potential of Exosomes d-MAPPS. Additionally, high concentrations of vascular endothelial growth factor receptor (VEGFR1) and chemokines (CXCL16, CCL21, CXCL14) were noticed at Exosomes d-MAPPS samples suggesting their potential to promote generation of new blood vessels and migration of CXCR6, CCR7 and CXCR4 expressing cells. Since all proteins which were found in high concentration in Exosomes d-MAPPS samples (IL-1Ra, CXCL16, CXCL14, CCL21, IL-27 and VEGFR1) are involved in modulation of lung, eye, and synovial inflammation, Exosomes d-MAPPS samples were prepared as inhalation and ophthalmic solutions in addition to injection formulations; their application in several patients suffering from chronic obstructive pulmonary disease, osteoarthritis, and dry eye syndrome resulted with significant improvement of biochemical and functional parameters. In conclusion, Exosomes d-MAPPS, due to the presence of important anti-inflammatory, immunomodulatory, and pro-angiogenic factors, represents potentially new therapeutic agent in regenerative medicine that should be further tested in large clinical studies.
Collapse
|
130
|
Li F, Li X, Liu G, Gao C, Li X. Bone Marrow Mesenchymal Stem Cells Decrease the Expression of RANKL in Collagen-Induced Arthritis Rats via Reducing the Levels of IL-22. J Immunol Res 2019; 2019:8459281. [PMID: 31828174 PMCID: PMC6885301 DOI: 10.1155/2019/8459281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To investigate the transplantation effect of bone marrow mesenchymal stem cells (MSCs) on the expression of interlukin-22 (IL-22) and RANKL in collagen-induced arthritis (CIA) rats. METHODS 32 CIA models were established. 16 CIA rats were transplanted with MSCs, and others were used as nontreatment CIA controls. The concentrations of IL-22 and RANKL in serum were detected by ELISA and those in synovial tissue of rats' joints by immunohistochemical staining. In addition, the expression of RANKL mRNA was measured by RT-PCR in the fibroblast-like synoviocytes (FLSs), cultured with IL-22 in vitro, which were delivered from the joints of CIA rats treated with or without MSCs. RESULTS The transplantation of MSCs into CIA rats relieved the destruction of joints, measured by AI score, X-ray, and histopathology. MSCs also reduced the expression of IL-22 and RANKL in serum by ELISA (P < 0.001) and similarly in FLSs by immunohistochemical staining. In vitro, IL-22 induced significantly the expression of RANKL mRNA in cultured FLSs in a dose-dependent manner, whereas this induction was significantly reduced in FLSs derived from CIA rats transplanted with MSCs (normal controls: F = 79.33, P < 0.001; CIA controls: F = 712.72, P < 0.001; and CIA-MSC rats: F = 139.04, P < 0.001). CONCLUSION Our results suggest that the transplantation of MSCs can reduce the expression of RANKL in vivo by downregulating the levels of IL-22, thereby ameliorating the degree of RA bone destruction. This study provides a theoretical basis for a potential therapy of RA with MSCs, and IL-22 and RANKL may become two new targets to treat RA.
Collapse
Affiliation(s)
- Fang Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xin Li
- Endocrine Metabolism and Immune Center, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Guiyan Liu
- Department of Nephrology, Changzhi People's Hospital, Changzhi 046000, China
| | - Chong Gao
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
131
|
Pinheiro D, Dias I, Ribeiro Silva K, Stumbo AC, Thole A, Cortez E, de Carvalho L, Weiskirchen R, Carvalho S. Mechanisms Underlying Cell Therapy in Liver Fibrosis: An Overview. Cells 2019; 8:cells8111339. [PMID: 31671842 PMCID: PMC6912561 DOI: 10.3390/cells8111339] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a common feature in most pathogenetic processes in the liver, and usually results from a chronic insult that depletes the regenerative capacity of hepatocytes and activates multiple inflammatory pathways, recruiting resident and circulating immune cells, endothelial cells, non-parenchymal hepatic stellate cells, and fibroblasts, which become activated and lead to excessive extracellular matrix accumulation. The ongoing development of liver fibrosis results in a clinically silent and progressive loss of hepatocyte function, demanding the constant need for liver transplantation in clinical practice, and motivating the search for other treatments as the chances of obtaining compatible viable livers become scarcer. Although initially cell therapy has emerged as a plausible alternative to organ transplantation, many factors still challenge the establishment of this technique as a main or even additional therapeutic tool. Herein, the authors discuss the most recent advances and point out the corners and some controversies over several protocols and models that have shown promising results as potential candidates for cell therapy for liver fibrosis, presenting the respective mechanisms proposed for liver regeneration in each case.
Collapse
Affiliation(s)
- Daphne Pinheiro
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Isabelle Dias
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Karina Ribeiro Silva
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Alessandra Thole
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Erika Cortez
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Lais de Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, D-52074 Aachen, Germany.
| | - Simone Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| |
Collapse
|
132
|
Dias IE, Pinto PO, Barros LC, Viegas CA, Dias IR, Carvalho PP. Mesenchymal stem cells therapy in companion animals: useful for immune-mediated diseases? BMC Vet Res 2019; 15:358. [PMID: 31640767 PMCID: PMC6805418 DOI: 10.1186/s12917-019-2087-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells are multipotent cells, with capacity for self-renewal and differentiation into tissues of mesodermal origin. These cells are possible therapeutic agents for autoimmune disorders, since they present remarkable immunomodulatory ability.The increase of immune-mediated diseases in veterinary medicine has led to a growing interest in the research of these disorders and their medical treatment. Conventional immunomodulatory drug therapy such as glucocorticoids or other novel therapies such as cyclosporine or monoclonal antibodies are associated with numerous side effects that limit its long-term use, leading to the need for developing new therapeutic strategies that can be more effective and safe.The aim of this review is to provide a critical overview about the therapeutic potential of these cells in the treatment of some autoimmune disorders (canine atopic dermatitis, feline chronic gingivostomatitis, inflammatory bowel disease and feline asthma) compared with their conventional treatment.Mesenchymal stem cell-based therapy in autoimmune diseases has been showing that this approach can ameliorate clinical signs or even cause remission in most animals, with the exception of canine atopic dermatitis in which little to no improvement was observed.Although mesenchymal stem cells present a promising future in the treatment of most of these disorders, the variability in the outcomes of some clinical trials has led to the current controversy among authors regarding their efficacy. Mesenchymal stem cell-based therapy is currently requiring a deeper and detailed analysis that allows its standardization and better adaptation to the intended therapeutic results, in order to overcome current limitations in future trials.
Collapse
Affiliation(s)
- Inês Esteves Dias
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
| | - Pedro Olivério Pinto
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
- Coimbra University Veterinary Hospital, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
| | - Luís Carlos Barros
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
| | - Carlos Antunes Viegas
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães Portugal
- ICVS/3B’s – PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Isabel Ribeiro Dias
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães Portugal
- ICVS/3B’s – PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Pedro Pires Carvalho
- CIVG - Vasco da Gama Research Center, Vasco da Gama University School, Av. José R. Sousa Fernandes 197, Campus Universitário - Bloco B, Lordemão, 3020-210 Coimbra, Portugal
- Vetherapy, 479 St, San Francisco, CA 94103 USA
| |
Collapse
|
133
|
Namba F. Mesenchymal stem cells for the prevention of bronchopulmonary dysplasia. Pediatr Int 2019; 61:945-950. [PMID: 31487104 DOI: 10.1111/ped.14001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/06/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in preterm infants who have been treated with supplemental oxygen and mechanical ventilation. Despite major advances in perinatal and neonatal medicine, limited progress has been made in reducing BPD rates. The use of mesenchymal stem cells (MSC) is a promising and innovative therapy for several diseases because they are easy to extract and they have low immunogenicity, anti-inflammatory properties, and regenerative ability. According to several pre-clinical studies that have used BPD animal models, one mechanism of action for MSC in BPD is mainly due to the paracrine effects of MSC-derived humoral factors, such as interleukin (IL)-6, IL-8, vascular endothelial growth factor, collagen, and elastin, rather than the multilineage and regenerative capacities of MSC. Cell-free preparations derived from MSC, including conditioned media and exosomes, remain a pre-clinical technology despite their great clinical potential. A first-in-human clinical trial of MSC treatment for BPD was performed as a phase I dose-escalation trial using umbilical cord blood-derived MSC. That trial demonstrated the short- and long-term safety and feasibility of MSC, given that significantly reduced inflammatory marker expression was observed in tracheal aspirates. As of recently, several clinical trials of MSC use for BPD are ongoing or are planned in some countries to investigate the efficacy of MSC in the prevention or treatment of BPD in premature infants. Many clinicians are currently awaiting the results from these trials so that MSC can be used clinically for human BPD.
Collapse
Affiliation(s)
- Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| |
Collapse
|
134
|
Abstract
Introduction: Bronchopulmonary dysplasia (BPD) is a common long-term adverse complication of very premature delivery. Affected infants can suffer chronic respiratory morbidities including lung function abnormalities and reduced exercise capacity even as young adults. Many studies have investigated possible preventative strategies; however, it is equally important to identify optimum management strategies for infants with evolving or established BPD. Areas covered: Respiratory support modalities and established and novel pharmacological treatments. Expert opinion: Respiratory support modalities including proportional assist ventilation and neurally adjusted ventilatory assist are associated with short term improvements in oxygenation indices. Such modalities need to be investigated in appropriate RCTs. Many pharmacological treatments are routinely used with a limited evidence base, for example diuretics. Stem cell therapies in small case series are associated with promising results. More research is required before it is possible to determine if such therapies should be investigated in large RCTs with long-term outcomes.
Collapse
Affiliation(s)
- Emma Williams
- a Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , UK.,b The Asthma UK Centre for Allergic Mechanisms in Asthma, King's College London , UK
| | - Anne Greenough
- a Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , UK.,c NIHR Biomedical Research Centre based at Guy's and St Thomas' NHS Foundation Trust and King's College London , London , UK
| |
Collapse
|
135
|
Lazarević JJ, Ralević U, Kukolj T, Bugarski D, Lazarević N, Bugarski B, Popović ZV. Influence of chemical fixation process on primary mesenchymal stem cells evidenced by Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 216:173-178. [PMID: 30897378 DOI: 10.1016/j.saa.2019.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 06/09/2023]
Abstract
In investigation of (patho)physiological processes, cells represent frequently used analyte as an exceptional source of information. However, spectroscopic analysis of live cells is still very seldom in clinics, as well as in research studies. Among others, the reasons are long acquisition time during which autolysis process is activated, necessity of specified technical equipment, and inability to perform analysis in a moment of sample preparation. Hence, an optimal method of preserving cells in the existing state is of extreme importance, having in mind that selection of fixative is cell lineage dependent. In this study, two commonly used chemical fixatives, formaldehyde and methanol, are used for preserving primary mesenchymal stem cells extracted from periodontal ligament, which are valuable cell source for reconstructive dentistry. By means of Raman spectroscopy, cell samples were probed and the impact of these fixatives on their Raman response was analyzed and compared. Different chemical mechanisms are the core processes of formaldehyde and methanol fixation and certain Raman bands are shifted and/or of changed intensity when Raman spectra of cells fixed in that manner are compared. In order to get clearer picture, comprehensive statistical analysis was performed.
Collapse
Affiliation(s)
- J J Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia
| | - U Ralević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia
| | - T Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - D Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - N Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia.
| | - B Bugarski
- Department of Chemical Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, Belgrade 11060, Serbia
| | - Z V Popović
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia; Serbian Academy of Sciences and Arts, Knez Mihailova 35, Belgrade 11000, Serbia
| |
Collapse
|
136
|
Lopez N, Ramamoorthy S, Sandborn WJ. Recent advances in the management of perianal fistulizing Crohn's disease: lessons for the clinic. Expert Rev Gastroenterol Hepatol 2019; 13:563-577. [PMID: 31023087 PMCID: PMC6545251 DOI: 10.1080/17474124.2019.1608818] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Managing fistulizing perianal disease is among the most challenging aspects of treating patients with Crohn's disease. Perianal fistulas are indicative of poor long-term prognosis. They are commonly associated with significant morbidities and can have detrimental effects on quality of life. While durable fistula closure is ideal, it is uncommon. In optimal circumstances, reported long-term fistula healing rates are only slightly higher than 50% and recurrence is common. Achieving these results requires a combined medical and surgical approach, highlighting the importance of a highly skilled and collaborative multidisciplinary team. In recent years, advances in imaging, biologic therapies and surgical techniques have lent to growing enthusiasm amongst treatment teams, however the most advantageous approach is yet to be determined. Areas covered: Here we review current management approaches, incorporating recent guidelines and novel therapies. Additionally, we discuss recently published and ongoing studies that will likely impact practice in the coming years. Expert opinion: Investing in concerted collaborative multi-institutional efforts will be necessary to better define optimal timing and dosing of medical therapy, as well as to identify ideal timing and approach of surgical interventions. Standardizing outcome measures can facilitate these efforts. Clearly, experienced multidisciplinary teams will be paramount in this process.
Collapse
Affiliation(s)
- Nicole Lopez
- Division of Colon and Rectal Surgery, University of California San Diego, California, USA
| | - Sonia Ramamoorthy
- Division of Colon and Rectal Surgery, University of California San Diego, California, USA
| | - Willam J. Sandborn
- Inflammatory Bowel Disease Center, University of California San Diego, California, USA,Division of Gastroenterology, University of California San Diego, California, USA
| |
Collapse
|
137
|
Harrell CR, Fellabaum C, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Molecular Mechanisms Responsible for Therapeutic Potential of Mesenchymal Stem Cell-Derived Secretome. Cells 2019; 8:cells8050467. [PMID: 31100966 PMCID: PMC6562906 DOI: 10.3390/cells8050467] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cell (MSC)-sourced secretome, defined as the set of MSC-derived bioactive factors (soluble proteins, nucleic acids, lipids and extracellular vesicles), showed therapeutic effects similar to those observed after transplantation of MSCs. MSC-derived secretome may bypass many side effects of MSC-based therapy, including unwanted differentiation of engrafted MSCs. In contrast to MSCs which had to be expanded in culture to reach optimal cell number for transplantation, MSC-sourced secretome is immediately available for treatment of acute conditions, including fulminant hepatitis, cerebral ischemia and myocardial infarction. Additionally, MSC-derived secretome could be massively produced from commercially available cell lines avoiding invasive cell collection procedure. In this review article we emphasized molecular and cellular mechanisms that were responsible for beneficial effects of MSC-derived secretomes in the treatment of degenerative and inflammatory diseases of hepatobiliary, respiratory, musculoskeletal, gastrointestinal, cardiovascular and nervous system. Results obtained in a large number of studies suggested that administration of MSC-derived secretomes represents a new, cell-free therapeutic approach for attenuation of inflammatory and degenerative diseases. Therapeutic effects of MSC-sourced secretomes relied on their capacity to deliver genetic material, growth and immunomodulatory factors to the target cells enabling activation of anti-apoptotic and pro-survival pathways that resulted in tissue repair and regeneration.
Collapse
Affiliation(s)
| | | | - Nemanja Jovicic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| | - Nebojsa Arsenijevic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| | - Vladislav Volarevic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| |
Collapse
|
138
|
Mesenchymal Stem Cell-Based Therapy of Inflammatory Lung Diseases: Current Understanding and Future Perspectives. Stem Cells Int 2019; 2019:4236973. [PMID: 31191672 PMCID: PMC6525794 DOI: 10.1155/2019/4236973] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/06/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
During acute or chronic lung injury, inappropriate immune response and/or aberrant repair process causes irreversible damage in lung tissue and most usually results in the development of fibrosis followed by decline in lung function. Inhaled corticosteroids and other anti-inflammatory drugs are very effective in patients with inflammatory lung disorders, but their long-term use is associated with severe side effects. Accordingly, new therapeutic agents that will attenuate ongoing inflammation and, at the same time, promote regeneration of injured alveolar epithelial cells are urgently needed. Mesenchymal stem cells (MSCs) are able to modulate proliferation, activation, and effector function of all immune cells that play an important role in the pathogenesis of acute and chronic inflammatory lung diseases. In addition to the suppression of lung-infiltrated immune cells, MSCs have potential to differentiate into alveolar epithelial cells in vitro and, accordingly, represent new players in cell-based therapy of inflammatory lung disorders. In this review article, we described molecular mechanisms involved in MSC-based therapy of acute and chronic pulmonary diseases and emphasized current knowledge and future perspectives related to the therapeutic application of MSCs in patients suffering from acute respiratory distress syndrome, pneumonia, asthma, chronic obstructive pulmonary diseases, and idiopathic pulmonary fibrosis.
Collapse
|
139
|
Xue Z, Ansari AR, Zhao X, Zang K, Liang Y, Cui L, Hu Y, Cheng R, Zhang X, Zhong J, Liu H. RNA-Seq-Based Gene Expression Pattern and Morphological Alterations in Chick Thymus during Postnatal Development. Int J Genomics 2019; 2019:6905194. [PMID: 31179312 PMCID: PMC6501151 DOI: 10.1155/2019/6905194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/20/2018] [Accepted: 02/25/2019] [Indexed: 12/23/2022] Open
Abstract
The thymus is a lobulated unique lymphoid immune organ that plays a critical role in the selection, development, proliferation, and differentiation of T cells. The thymus of developing chickens undergoes continued morphological alterations; however, the biomolecular and transcriptional dynamics of the postnatal thymus in avian species is not clear yet. Therefore, the thymuses from chickens at different stages of development (at weeks 0, 1, 5, 9, 18, and 27) were used in the present study. The RNA-seq method was used to study the gene expression patterns. On average, 24120819 clean reads were mapped, differentially expressed genes (DEGs) were identified on the basis of log values (fold change), including 744 upregulated and 425 downregulated genes. The expression pattern revealed by RNA-seq was validated by quantitative real-time PCR (qPCR) analysis of four important genes, which are PCNA, CCNA2, CCNB2, and CDK1. Thus, the current study revealed that during postnatal development, the thymus undergoes severe atrophy. Thymus structure was damaged and gene expression changed dramatically, especially at the 27th week of age. Moreover, we found significant changes of several signaling pathways such as the cytokine-cytokine receptor interaction and cell cycle signaling pathways. Hence, it may be inferred that those signaling pathways might be closely related to the postnatal chicken thymus development.
Collapse
Affiliation(s)
- Zhouyiyuan Xue
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Abdur Rahman Ansari
- Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS) Jhang, University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Xing Zhao
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Kun Zang
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yu Liang
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Lei Cui
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yafang Hu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ranran Cheng
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiaolong Zhang
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Juming Zhong
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, USA
| | - Huazhen Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
140
|
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: 46] [Impact Index Per Article: 7.7] [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
|
141
|
Lazarević JJ, Kukolj T, Bugarski D, Lazarević N, Bugarski B, Popović ZV. Probing primary mesenchymal stem cells differentiation status by micro-Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 213:384-390. [PMID: 30726762 DOI: 10.1016/j.saa.2019.01.069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/21/2019] [Indexed: 05/27/2023]
Abstract
We have employed micro-Raman spectroscopy to get insight into intrinsic biomolecular profile of individual mesenchymal stem cell isolated from periodontal ligament. Furthermore, these cells were stimulated towards adipogenic, chondrogenic, and osteogenic lineages and their status of differentiation was assessed using micro-Raman spectroscopy. In both cases, glass coverslips were used as substrates, due to their wide availability and cost effectiveness. In all sample groups, the same type of behavior was observed, manifested as changes in Raman spectra: the increase of relative intensity of protein/lipid bands and decrease of nucleic acid bands. Comprehensive statistical analysis in the form of principal component analysis was performed, which revealed noticeable grouping of cells with the similar features. Despite the inhomogeneity of primary stem cells and their differentiated lineages, we demonstrated that micro-Raman spectroscopy is sufficient for distinguishing cells' status, which can be valuable for medical and clinical application.
Collapse
Affiliation(s)
- J J Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia
| | - T Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - D Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - N Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia.
| | - B Bugarski
- Department of Chemical Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, Belgrade 11060, Serbia
| | - Z V Popović
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia; Serbian Academy of Sciences and Arts, Knez Mihailova 35, Belgrade 11000, Serbia
| |
Collapse
|
142
|
Mucientes A, Herranz E, Moro E, Lajas C, Candelas G, Fernández-Gutiérrez B, Lamas JR. Differential Expression of HOX Genes in Mesenchymal Stem Cells from Osteoarthritic Patients Is Independent of Their Promoter Methylation. Cells 2018; 7:cells7120244. [PMID: 30563049 PMCID: PMC6316585 DOI: 10.3390/cells7120244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022] Open
Abstract
Skeletogenesis, remodeling, and maintenance in adult tissues are regulated by sequential activation of genes coding for specific transcription factors. The conserved Homeobox genes (HOX, in humans) are involved in several skeletal pathologies. Osteoarthritis (OA) is characterized by homeostatic alterations of cartilage and bone synthesis, resulting in cartilage destruction and increased bone formation. We postulate that alterations in HOX expression in Mesenchymal Stem cells (MSCs) are likely one of the causes explaining the homeostatic alterations in OA and that this altered expression could be the result of epigenetic regulation. The expression of HOX genes in osteoarthritic-derived MSCs was screened using PCR arrays. Epigenetic regulation of HOX was analyzed measuring the degree of DNA methylation in their promoters. We demonstrate the downregulated expression of HOXA9 and HOXC8 in OA-MSCs. However, their expression does not correlate with promoter methylation status, suggesting that other epigenetic mechanisms could be implicated in the regulation of HOX expression. Studies on the role of these genes under active differentiation conditions need to be addressed for a better knowledge of the mechanisms regulating the expression of HOX, to allow a better understanding of OA pathology and to define possible biomarkers for therapeutic treatment.
Collapse
Affiliation(s)
- Arkaitz Mucientes
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - Eva Herranz
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - Enrique Moro
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Traumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - Cristina Lajas
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - Gloria Candelas
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - Benjamín Fernández-Gutiérrez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| | - José Ramón Lamas
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC). UGC de Reumatología, Hospital Clínico San Carlos, 28040 Madrid, Spain.
| |
Collapse
|
143
|
Villatoro AJ, Hermida-Prieto M, Fernández V, Fariñas F, Alcoholado C, Rodríguez-García MI, Mariñas-Pardo L, Becerra J. Allogeneic adipose-derived mesenchymal stem cell therapy in dogs with refractory atopic dermatitis: clinical efficacy and safety. Vet Rec 2018; 183:654. [DOI: 10.1136/vr.104867] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/18/2018] [Accepted: 08/05/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Antonio José Villatoro
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences; University of Málaga, IBIMA; Málaga Spain
- Cellular Therapy Unit; Instituto de Inmunología Clínica y Terapia Celular (IMMUNESTEM); Málaga Spain
| | | | - Viviana Fernández
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences; University of Málaga, IBIMA; Málaga Spain
- Cellular Therapy Unit; Instituto de Inmunología Clínica y Terapia Celular (IMMUNESTEM); Málaga Spain
| | - Fernando Fariñas
- Cellular Therapy Unit; Instituto de Inmunología Clínica y Terapia Celular (IMMUNESTEM); Málaga Spain
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences; University of Málaga, IBIMA; Málaga Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Madrid Spain
| | | | | | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences; University of Málaga, IBIMA; Málaga Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Madrid Spain
- Andalusian Centre for Nanomedicine and Biotechnology-BIONAND; Málaga Spain
| |
Collapse
|
144
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med 2018; 23:720-730. [PMID: 30484934 PMCID: PMC6349184 DOI: 10.1111/jcmm.14035] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common, severe emergency case in clinics, with high incidence, significant mortality and increased costs. Despite development in the understanding of its pathophysiology, the therapeutic choices are still confined to dialysis and renal transplantation. Considering their antiapoptotic, immunomodulatory, antioxidative and pro‐angiogenic effects, mesenchymal stem cells (MSCs) may be a promising candidate for AKI management. Based on these findings, some clinical trials have been performed, but the results are contradictory (NCT00733876, NCT01602328). The low engraftment, poor survival rate, impaired paracrine ability and delayed administration of MSCs are the four main reasons for the limited clinical efficacy. Investigators have developed a series of preconditioning strategies to improve MSC survival rates and paracrine ability. In this review, by summarizing these encouraging studies, we intend to provide a comprehensive understanding of various preconditioning strategies on AKI therapy and improve the prognosis of AKI patients by regenerative medicine.
Collapse
Affiliation(s)
- Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Zhang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hua Jiang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
145
|
Clinical Application of Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapeutics for Inflammatory Lung Diseases. J Clin Med 2018; 7:jcm7100355. [PMID: 30322213 PMCID: PMC6210470 DOI: 10.3390/jcm7100355] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/01/2018] [Accepted: 10/12/2018] [Indexed: 02/06/2023] Open
Abstract
It is currently thought that extracellular vesicles (EVs), such as exosomes and microvesicles, play an important autocrine/paracrine role in intercellular communication. EVs package proteins, mRNA and microRNA (miRNA), which have the ability to transfer biological information to recipient cells in the lungs. Depending on their origin, EVs fulfil different functions. EVs derived from mesenchymal stem cells (MSCs) have been found to promote therapeutic activities that are comparable to MSCs themselves. Recent animal model-based studies suggest that MSC-derived EVs have significant potential as a novel alternative to whole-cell therapies. Compared to their parent cells, EVs may have a superior safety profile and can be stored without losing function. It has been observed that MSC-derived EVs suppress pro-inflammatory processes and reduce oxidative stress, pulmonary fibrosis and remodeling in a variety of in vivo inflammatory lung disease models by transferring their components. However, there remain significant challenges to translate this therapy to the clinic. From this view point, we will summarize recent studies on EVs produced by MSCs in preclinical experimental models of inflammatory lung diseases. We will also discuss the most relevant issues in bringing MSC-derived EV-based therapeutics to the clinic for the treatment of inflammatory lung diseases.
Collapse
|
146
|
|
147
|
Jakovljevic J, Harrell CR, Fellabaum C, Arsenijevic A, Jovicic N, Volarevic V. Modulation of autophagy as new approach in mesenchymal stem cell-based therapy. Biomed Pharmacother 2018; 104:404-410. [PMID: 29787987 DOI: 10.1016/j.biopha.2018.05.061] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Due to their trophic and immunoregulatory characteristics mesenchymal stem cells (MSCs) have tremendous potential for use in a variety of clinical applications. Challenges in MSCs' clinical applications include low survival of transplanted cells and low grafting efficiency requiring use of a high number of MSCs to achieve therapeutic benefits. Accordingly, new approaches are urgently needed in order to overcome these limitations. Recent evidence indicates that modulation of autophagy in MSCs prior to their transplantation enhances survival and viability of engrafted MSCs and promotes their pro-angiogenic and immunomodulatory characteristics. Here, we review the current literature describing mechanisms by which modulation of autophagy strengthens pro-angiogenic and immunosuppressive characteristics of MSCs in animal models of multiple sclerosis, osteoporosis, diabetic limb ischemia, myocardial infarction, acute graft-versus-host disease, kidney and liver diseases. Obtained results suggest that modulation of autophagy in MSCs may represent a new therapeutic approach that could enhance efficacy of MSCs in the treatment of ischemic and autoimmune diseases.
Collapse
Affiliation(s)
- Jelena Jakovljevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - C Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida, United States
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida, United States
| | - Aleksandar Arsenijevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - Nemanja Jovicic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - Vladislav Volarevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia.
| |
Collapse
|
148
|
Gazdic M, Markovic BS, Arsenijevic A, Jovicic N, Acovic A, Harrell CR, Fellabaum C, Djonov V, Arsenijevic N, Lukic ML, Volarevic V. Crosstalk between mesenchymal stem cells and T regulatory cells is crucially important for the attenuation of acute liver injury. Liver Transpl 2018; 24:687-702. [PMID: 29500914 DOI: 10.1002/lt.25049] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 12/15/2022]
Abstract
One of the therapeutic options for the treatment of fulminant hepatitis is repopulation of intrahepatic regulatory cells because their pool is significantly reduced during acute liver failure. Although it is known that mesenchymal stem cells (MSCs), which have beneficent effects in the therapy of fulminant hepatitis, may promote expansion of regulatory T cells (Tregs) and regulatory B cells (Bregs), the role of these regulatory cells in MSC-mediated attenuation of acute liver injury is unknown. Herewith, we described the molecular mechanisms involved in the crosstalk between MSCs and liver regulatory cells and analyzed the potential of MSC-based therapy for the expansion of intrahepatic regulatory cells in mouse model of acute liver failure. MSC-dependent attenuation of α-galactosylceramide (α-GalCer)-induced acute liver injury in mice was accompanied with an increased presence of interleukin (IL) 10-producing CD4+ CD25+ forkhead box P3+ Tregs and IL10- and transforming growth factor β-producing marginal zone-like Bregs in the liver. Depletion of Bregs did not alter MSC-based alleviation of acute liver failure, whereas depletion of Tregs completely abrogated hepatoprotective effects of MSCs and inhibited their capacity to attenuate hepatotoxicity of liver natural killer T cells (NKTs), indicating that Tregs, and not Bregs, were critically involved in MSC-based modulation of acute liver inflammation. MSCs, in a paracrine, indoleamine 2,3-dioxygenase-dependent manner, significantly increased the capacity of Tregs to produce immunosuppressive IL10 and to suppress hepatotoxicity of liver NKTs. Accordingly, adoptive transfer of MSC-primed Tregs resulted in the complete attenuation of α-GalCer-induced acute liver failure. In conclusion, our findings highlighted the crucial importance of Tregs for MSC-based attenuation of acute liver failure and indicated the significance of MSC-mediated priming of Tregs as a new therapeutic approach in Treg-based therapy of acute liver injury. Liver Transplantation 24 687-702 2018 AASLD.
Collapse
Affiliation(s)
- Marina Gazdic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Simovic Markovic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Acovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | | | - Nebojsa Arsenijevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladislav Volarevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
149
|
Villatoro AJ, Claros S, Fernández V, Alcoholado C, Fariñas F, Moreno A, Becerra J, Andrades JA. Safety and efficacy of the mesenchymal stem cell in feline eosinophilic keratitis treatment. BMC Vet Res 2018; 14:116. [PMID: 29587744 PMCID: PMC5870249 DOI: 10.1186/s12917-018-1413-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/06/2018] [Indexed: 01/07/2023] Open
Abstract
Background Feline eosinophilic keratitis (FEK) is a chronic keratopathy caused by a suspected immune mediated response to an unknown antigenic stimulus. The purpose of this study was to investigate the safety and therapeutic effects of allogeneic feline adipose-derived mesenchymal stromal cells (fAd-MSCs) implanted subconjunctival around the ocular surface lesion in five cats with FEK refractory to current available treatments. Results FEK was diagnosed by clinical appearance and evidence of eosinophil and/or mast cells in corneal cytology. Each animal was treated with two applications of 2 × 106 million of fAd-MSCs 2 months apart. Ocular surface integrity was assessed before treatment and at 1, 3, 6 and 11 months after treatment. Clinical signs showed a significant change during the follow-up with resolution of the corneal and conjunctiva lesions and there were no signs of regression or worsening. Conclusions Implanted cells were well-tolerated and effective reducing clinical signs of FEK with a sustained effect during the study period. None of the animals showed systemic or local complications during the study. To our knowledge, this is the first time in literature that local implantation of allogeneic fAd-MSCs has been found as an effective therapeutic alternative to treat cats with FEK.
Collapse
Affiliation(s)
- Antonio J Villatoro
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Silvia Claros
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Viviana Fernández
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Fernando Fariñas
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Antonio Moreno
- Hospital veterinario Alhaurín el Grande. Alhaurín el Grande, 29120, Málaga, Spain
| | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.,Laboratory of Bioengineering and Tissue Regeneration, Andalusian Center for Nanomedicine and Biotechnology-BIONAND, 29590, Málaga, Spain
| | - José A Andrades
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain. .,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.
| |
Collapse
|
150
|
Nikolic A, Simovic Markovic B, Gazdic M, Randall Harrell C, Fellabaum C, Jovicic N, Djonov V, Arsenijevic N, L Lukic M, Stojkovic M, Volarevic V. Intraperitoneal administration of mesenchymal stem cells ameliorates acute dextran sulfate sodium-induced colitis by suppressing dendritic cells. Biomed Pharmacother 2018; 100:426-432. [PMID: 29471245 DOI: 10.1016/j.biopha.2018.02.060] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/02/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) have important pathogenic role in the induction and progression of ulcerative colitis (UC), but their role in mesenchymal stem cells (MSCs)-mediated suppression of colon injury and inflammation is not revealed. By using dextran sodium sulfate (DSS)-induced colitis, a well-established murine model of UC, we examined effects of MSCs on phenotype and function of colon infiltrating DCs. Clinical, histological, immunophenotypic analysis and passive transfer of MSCs-primed DCs were used to evaluate capacity of MSC to suppress inflammatory phenotype of DCs in vivo. Additionally, DCs:MSCs interplay was also investigated in vitro, to confirmed in vivo obtained findings. Intraperitoneally administered MSCs (2 × 106) significantly reduced progression of DSS-induced colitis and reduced serum levels of inflammatory cytokines (IL-1β, IL-12, and IL-6). Passive transfer of in vivo MSCs-primed DCs reduced severity of colitis while passive transfer of MSCs-non-primed DCs aggravated DSS-induced colitis. Through the secretion of immunomodulatory Galectin 3, MSCs, in paracrine manner, down-regulated production of inflammatory cytokines in DCs and attenuated expression of co-stimulatory and major histocompatibility complex class II molecules on their membranes. Taken together, these results indicate that MSCs achieved their beneficial effects in DSS-induced colitis by suppressing inflammatory phenotype of DCs in Gal-3 dependent manner. Therapeutic targeting of DCs by MSCs should be explored in future studies as a useful approach for the treatment of UC.
Collapse
Affiliation(s)
- Aleksandar Nikolic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Simovic Markovic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - C Randall Harrell
- Regenerative Processing Plant, LLC, Palm Harbor, Florida, United States
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, Palm Harbor, Florida, United States
| | - Nemanja Jovicic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Nebojsa Arsenijevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag Stojkovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; Spebo Medical, Leskovac, Serbia
| | - Vladislav Volarevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| |
Collapse
|