1
|
Pharoun J, Berro J, Sobh J, Abou-Younes MM, Nasr L, Majed A, Khalil A, Joseph, Stephan, Faour WH. Mesenchymal stem cells biological and biotechnological advances: Implications for clinical applications. Eur J Pharmacol 2024; 977:176719. [PMID: 38849038 DOI: 10.1016/j.ejphar.2024.176719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into multiple lineages including bone, cartilage, muscle and fat. They hold immunomodulatory properties and therapeutic ability to treat multiple diseases, including autoimmune and chronic degenerative diseases. In this article, we reviewed the different biological properties, applications and clinical trials of MSCs. Also, we discussed the basics of manufacturing conditions, quality control, and challenges facing MSCs in the clinical setting. METHODS Extensive review of the literature was conducted through the databases PubMed, Google Scholar, and Cochrane. Papers published since 2015 and covering the clinical applications and research of MSC therapy were considered. Furthermore, older papers were considered when referring to pioneering studies in the field. RESULTS The most widely studied stem cells in cell therapy and tissue repair are bone marrow-derived mesenchymal stem cells. Adipose tissue-derived stem cells became more common and to a lesser extent other stem cell sources e.g., foreskin derived MSCs. MSCs therapy were also studied in the setting of COVID-19 infections, ischemic strokes, autoimmune diseases, tumor development and graft rejection. Multiple obstacles, still face the standardization and optimization of MSC therapy such as the survival and the immunophenotype and the efficiency of transplanted cells. MSCs used in clinical settings displayed heterogeneity in their function despite their extraction from healthy donors and expression of similar surface markers. CONCLUSION Mesenchymal stem cells offer a rising therapeutic promise in various diseases. However, their potential use in clinical applications requires further investigation.
Collapse
Affiliation(s)
- Jana Pharoun
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jana Berro
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jeanine Sobh
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | | | - Leah Nasr
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Ali Majed
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Alia Khalil
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Joseph
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Stephan
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36.
| |
Collapse
|
2
|
Kwon HY, Yoon Y, Hong JE, Rhee KJ, Sohn JH, Jung PY, Kim MY, Baik SK, Ryu H, Eom YW. Role of TGF-β and p38 MAPK in TSG-6 Expression in Adipose Tissue-Derived Stem Cells In Vitro and In Vivo. Int J Mol Sci 2023; 25:477. [PMID: 38203646 PMCID: PMC10778696 DOI: 10.3390/ijms25010477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Mesenchymal stem cells (MSCs) regulate immune cell activity by expressing tumor necrosis factor-α (TNF-α)-stimulated gene 6 (TSG-6) in inflammatory environments; however, whether anti-inflammatory responses affect TSG-6 expression in MSCs is not well understood. Therefore, we investigated whether transforming growth factor-β (TGF-β) regulates TSG-6 expression in adipose tissue-derived stem cells (ASCs) and whether effective immunosuppression can be achieved using ASCs and TGF-β signaling inhibitor A83-01. TGF-β significantly decreased TSG-6 expression in ASCs, but A83-01 and the p38 inhibitor SB202190 significantly increased it. However, in septic C57BL/6 mice, A83-01 further reduced the survival rate of the lipopolysaccharide (LPS)-treated group and ASC transplantation did not improve the severity induced by LPS. ASC transplantation alleviated the severity of sepsis induced by LPS+A83-01. In co-culture of macrophages and ASCs, A83-01 decreased TSG-6 expression whereas A83-01 and SB202190 reduced Cox-2 and IDO-2 expression in ASCs. These results suggest that TSG-6 expression in ASCs can be regulated by high concentrations of pro-inflammatory cytokines in vitro and in vivo, and that A83-01 and SB202190 can reduce the expression of immunomodulators in ASCs. Therefore, our data suggest that co-treatment of ASCs with TGF-β or p38 inhibitors is not adequate to modulate inflammation.
Collapse
Affiliation(s)
- Hye Youn Kwon
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Yongdae Yoon
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (J.-E.H.); (K.-J.R.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (J.-E.H.); (K.-J.R.)
| | - Joon Hyung Sohn
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| | - Pil Young Jung
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Moon Young Kim
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Soon Koo Baik
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Hoon Ryu
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (H.Y.K.); (P.Y.J.)
| | - Young Woo Eom
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea; (Y.Y.); (M.Y.K.); (S.K.B.)
| |
Collapse
|
3
|
Wu J, Huang QM, Liu Y, Zhou J, Tang WR, Wang XY, Wang LF, Zhang ZH, Tan HL, Guan XH, Deng KY, Xin HB. Long-term hypoxic hUCMSCs-derived extracellular vesicles alleviates allergic rhinitis through triggering immunotolerance of their VEGF-mediated inhibition of dendritic cells maturation. Int Immunopharmacol 2023; 124:110875. [PMID: 37742368 DOI: 10.1016/j.intimp.2023.110875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/18/2023] [Accepted: 08/27/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Extensions of mesenchymal stem cells (MSCs) in vitro may lead to the loss of their biological functions. However, hypoxic culturation has been shown to enhance the proliferation, survival, and immunomodulatory capacity of MSCs. OBJECTIVE We aimed to investigate the effects of long-term hypoxic cultivation on the properties of human umbilical cord-derived MSCs (hUCMSCs) and the therapeutic effects of their extracellular vesicles (EVs) in allergic rhinitis (AR). METHODS Proliferation, senescence, telomerase activity and multipotent properties of hUCMSCs were analyzed under long-term culturation of hypoxia (1%) or normoxia (21%), and the therapeutic effects of their conditional medium (CM) and EVs were evaluated in OVA-induced AR mice. Effects of hypoxia-EVs (Hy-EVs) or normoxia-EVs (No-EVs) on human monocyte-derived dendritic cells (DCs) were investigated, and the possible mechanisms of Hy-EVs in induction of immunotolerance were further explored. RESULTS Long-term hypoxia significantly promoted the proliferation, inhibited cell senescence, maintained the multipotent status of hUCMSCs. Hy-CM and Hy-EVs showed better therapeutic effects in AR mice compared to No-EVs, seen as improvement of AR-related behaviors such as rubbing and sneezing, and attenuation of inflammation in nasal tissues. In addition, Hy-EVs significantly reduced the expressions of HLA-DR, CD80, CD40, and CD83 induced by OVA plus LPS in DCs, inhibiting the maturation of DCs. Furthermore, we observed that VEGF was remarkably enriched in Hy-EVs, but not in No-EVs, and the inhibition of DCs maturation was markedly neutralized by VEGF antibodies, suggesting that VEGF derived from Hy-EVs was responsible for the inhibition of DCs maturation. CONCLUSION Our results demonstrated that long-term hypoxia significantly promoted the proliferation, inhibited cell senescence, maintained the multipotent status of hUCMSCs, and hypoxia treated hUCMSCs-derived EVs enhanced their therapeutic effects in AR mice through VEGF-mediated inhibition of DCs maturation.
Collapse
Affiliation(s)
- Jie Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; College of Life Science, Nanchang University, Nanchang 330031, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330052, China
| | - Qi-Ming Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; College of Life Science, Nanchang University, Nanchang 330031, China
| | - Yu Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330052, China
| | - Juan Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang 330052, China
| | - Wen-Rong Tang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Xiao-Yu Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Lin-Fang Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Zhou-Hang Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Hui-Lan Tan
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Xiao-Hui Guan
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China.
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; College of Life Science, Nanchang University, Nanchang 330031, China.
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; College of Life Science, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
4
|
Hawthorne IJ, Dunbar H, Tunstead C, Schorpp T, Weiss DJ, Enes SR, Dos Santos CC, Armstrong ME, Donnelly SC, English K. Human macrophage migration inhibitory factor potentiates mesenchymal stromal cell efficacy in a clinically relevant model of allergic asthma. Mol Ther 2023; 31:3243-3258. [PMID: 37735872 PMCID: PMC10638061 DOI: 10.1016/j.ymthe.2023.09.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
Current asthma therapies focus on reducing symptoms but fail to restore existing structural damage. Mesenchymal stromal cell (MSC) administration can ameliorate airway inflammation and reverse airway remodeling. However, differences in patient disease microenvironments seem to influence MSC therapeutic effects. A polymorphic CATT tetranucleotide repeat at position 794 of the human macrophage migration inhibitory factor (hMIF) gene has been associated with increased susceptibility to and severity of asthma. We investigated the efficacy of human MSCs in high- vs. low-hMIF environments and the impact of MIF pre-licensing of MSCs using humanized MIF mice in a clinically relevant house dust mite (HDM) model of allergic asthma. MSCs significantly attenuated airway inflammation and airway remodeling in high-MIF-expressing CATT7 mice but not in CATT5 or wild-type littermates. Differences in efficacy were correlated with increased MSC retention in the lungs of CATT7 mice. MIF licensing potentiated MSC anti-inflammatory effects at a previously ineffective dose. Mechanistically, MIF binding to CD74 expressed on MSCs leads to upregulation of cyclooxygenase 2 (COX-2) expression. Blockade of CD74 or COX-2 function in MSCs prior to administration attenuated the efficacy of MIF-licensed MSCs in vivo. These findings suggest that MSC administration may be more efficacious in severe asthma patients with high MIF genotypes (CATT6/7/8).
Collapse
Affiliation(s)
- Ian J Hawthorne
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland; Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Hazel Dunbar
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland; Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Courteney Tunstead
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland; Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Tamara Schorpp
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland; Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Daniel J Weiss
- Department of Medicine, 226 Health Sciences Research Facility, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, 30 Bond Street, Toronto, ON, Canada; Institute of Medical Sciences and Interdepartmental Division of Critical Care, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | | | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland; Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
5
|
Peng YQ, Deng XH, Xu ZB, Wu ZC, Fu QL. Mesenchymal stromal cells and their small extracellular vesicles in allergic diseases: From immunomodulation to therapy. Eur J Immunol 2023; 53:e2149510. [PMID: 37572379 DOI: 10.1002/eji.202149510] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/09/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
Mesenchymal stromal cells (MSCs) have long been considered a potential tool for treatment of allergic inflammatory diseases, owing to their immunomodulatory characteristics. In recent decades, the medical utility of MSCs has been evaluated both in vitro and in vivo, providing a foundation for therapeutic applications. However, the existing limitations of MSC therapy indicate the necessity for novel therapies. Notably, small extracellular vesicles (sEV) derived from MSCs have emerged rapidly as candidates instead of their parental cells. The acquisition of abundant and scalable MSC-sEV is an obstacle for clinical applications. The potential application of MSC-sEV in allergic diseases has attracted increasing attention from researchers. By carrying biological microRNAs or active proteins, MSC-sEV can modulate the function of various innate and adaptive immune cells. In this review, we summarise the recent advances in the immunomodulatory properties of MSCs in allergic diseases, the cellular sources of MSC-sEV, and the methods for obtaining high-quality human MSC-sEV. In addition, we discuss the immunoregulatory capacity of MSCs and MSC-sEV for the treatment of asthma, atopic dermatitis, and allergic rhinitis, with a special emphasis on their immunoregulatory effects and the underlying mechanisms of immune cell modulation.
Collapse
Affiliation(s)
- Ya-Qi Peng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiao-Hui Deng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Bin Xu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zi-Cong Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Hernandez Pichardo A, Wilm B, Liptrott NJ, Murray P. Intravenous Administration of Human Umbilical Cord Mesenchymal Stromal Cells Leads to an Inflammatory Response in the Lung. Stem Cells Int 2023; 2023:7397819. [PMID: 37705699 PMCID: PMC10497368 DOI: 10.1155/2023/7397819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/25/2023] [Accepted: 08/04/2023] [Indexed: 09/15/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) administered intravenously (IV) have shown efficacy in preclinical models of various diseases. This is despite the cells not reaching the site of injury due to entrapment in the lungs. The immunomodulatory properties of MSCs are thought to underlie their therapeutic effects, irrespective of whether they are sourced from bone marrow, adipose tissue, or umbilical cord. To better understand how MSCs affect innate immune cell populations in the lung, we evaluated the distribution and phenotype of neutrophils, monocytes, and macrophages by flow cytometry and histological analyses after delivering human umbilical cord-derived MSCs (hUC-MSCs) IV into immunocompetent mice. After 2 hr, we observed a significant increase in neutrophils, and proinflammatory monocytes and macrophages. Moreover, these immune cells localized in close proximity to the MSCs, suggesting an active role in their clearance. By 24 hr, we detected an increase in anti-inflammatory monocytes and macrophages. These results suggest that the IV injection of hUC-MSCs leads to an initial inflammatory phase in the lung shortly after injection, followed by a resolution phase 24 hr later.
Collapse
Affiliation(s)
- Alejandra Hernandez Pichardo
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Neill J. Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Pre-Clinical Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
7
|
Gholami M, Ghorban K, Sadeghi M, Dadmanesh M, Rouzbahani NH, Dehnavi S. Mesenchymal stem cells and allergic airway inflammation; a therapeutic approach to induce immunoregulatory responses. Int Immunopharmacol 2023; 120:110367. [PMID: 37230032 DOI: 10.1016/j.intimp.2023.110367] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/07/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Allergic airway inflammations are among the essential disorders worldwide that are already considered a significant concern. Mesenchymal stem cells (MSCs) are stromal cells with regenerative potential and immunomodulatory characteristics and are widely administered for tissue repair as an immunoregulatory agent in different inflammatory diseases. The current review summarized primary studies conducted to evaluate the therapeutic potential of MSCs for allergic airway disorders. In this case, modulation of airway pathologic inflammation and infiltration of inflammatory cells were examined, and modulation of the Th1/Th2 cellular balance and humoral responses. Also, the effects of MSCs on the Th17/Treg ratio and inducing Treg immunoregulatory responses along with macrophage and dendritic cell function were evaluated.
Collapse
Affiliation(s)
- Mohammad Gholami
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Medical Microbiology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Khodayar Ghorban
- Department of Immunology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Dadmanesh
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, School Of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Negin Hosseini Rouzbahani
- Infectious Diseases Research Center, Aja University of Medical Sciences, Tehran, Iran; Department of Immunology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Razi S, Yaghmoorian Khojini J, Kargarijam F, Panahi S, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Macrophage efferocytosis in health and disease. Cell Biochem Funct 2023; 41:152-165. [PMID: 36794573 DOI: 10.1002/cbf.3780] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Creating cellular homeostasis within a defined tissue typically relates to the processes of apoptosis and efferocytosis. A great example here is cell debris that must be removed to prevent unwanted inflammatory responses and then reduce autoimmunity. In view of that, defective efferocytosis is often assumed to be responsible for the improper clearance of apoptotic cells (ACs). This predicament triggers off inflammation and even results in disease development. Any disruption of phagocytic receptors, molecules as bridging groups, or signaling routes can also inhibit macrophage efferocytosis and lead to the impaired clearance of the apoptotic body. In this line, macrophages as professional phagocytic cells take the lead in the efferocytosis process. As well, insufficiency in macrophage efferocytosis facilitates the spread of a wide variety of diseases, including neurodegenerative diseases, kidney problems, types of cancer, asthma, and the like. Establishing the functions of macrophages in this respect can be thus useful in the treatment of many diseases. Against this background, this review aimed to recapitulate the knowledge about the mechanisms related to macrophage polarization under physiological or pathological conditions, and shed light on its interaction with efferocytosis.
Collapse
Affiliation(s)
- Shokufeh Razi
- Department of Genetics, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Kargarijam
- Department of Biotechnology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran
| | - Susan Panahi
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
9
|
Wang J, Zhang A, Huang F, Xu J, Zhao M. MSC-EXO and tempol ameliorate bronchopulmonary dysplasia in newborn rats by activating HIF-1α. Pediatr Pulmonol 2023; 58:1367-1379. [PMID: 36650825 DOI: 10.1002/ppul.26317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 12/25/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a major complication of premature infants and an important cause of morbidity and mortality. This study investigates the effect of the combination of mesenchymal stem cells-derived exosomes (MSC-EXO) and tempol on BPD and analyzes its mechanism. METHODS MSC-EXO was extracted by centrifugation and identified by transmission electron microscopy (TEM), nanoparticle tracking analysis, and western blot analysis (WB). Tidal volume (TV), minute ventilation (MV), peak inspiratory flow (PIF), and dynamic pulmonary compliance (Cdyn) of rats were measured by BuxCo pulmonary function experimental platform. Hematoxylin-eosin staining was performed to observe the lung morphology and radical alveolar count (RAC) and mean linear intercept (MLI) were assessed. Immunofluorescence (IF) was conducted to detect the expression of CD31 and α-SMA in pulmonary blood vessels. The kits were used to calculate malondialdehyde (MDA), superoxide dismutase (SOD), and total antioxidant capacity (TAOC) concentration in lung tissue. Enzyme linked immunosorbent assay was applied to detect the levels of IL-1β, IL-17, IL-6, and IFN-γ in bronchoalveolar lavage fluid. In addition, the expressions of HIF-1α, vascular endothelial growth factor (VEGF), p-PI3K, and p-AKT were analyzed by WB and IF. RESULTS We successfully extracted and identified MSC-EXO. In BPD rats, TV, MV, PIF, and Cdyn decreased, alveoli were simplified, and the number of interalveoli small vessels, blood vessel density decreased. Moreover, RAC, CD31, TAOC, and SOD decreased, and MLI, α-SMA, MDA, IL-1β, IL-17, IL-6, and IFN-γ increased, which was reversed by the combination of MSC-EXO and tempol treatment after combined treatment. In addition, the expression levels of HIF-1α, VEGF, p-PI3K, and p-AKT were increased after combined treatment. CONCLUSIONS Combined treatment could improve lung tissue injury, promote pulmonary vascular remodeling, restore lung function, and inhibit oxidative stress in BPD rats. These effects were achieved through activation of HIF-1α.
Collapse
Affiliation(s)
- Juanmei Wang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China.,Hunan Provincial Key Laboratory of Pediatric Respirology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Aimin Zhang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Furong Huang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Jun Xu
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Menghua Zhao
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
10
|
Boland L, Bitterlich LM, Hogan AE, Ankrum JA, English K. Translating MSC Therapy in the Age of Obesity. Front Immunol 2022; 13:943333. [PMID: 35860241 PMCID: PMC9289617 DOI: 10.3389/fimmu.2022.943333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/10/2022] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal cell (MSC) therapy has seen increased attention as a possible option to treat a number of inflammatory conditions including COVID-19 acute respiratory distress syndrome (ARDS). As rates of obesity and metabolic disease continue to rise worldwide, increasing proportions of patients treated with MSC therapy will be living with obesity. The obese environment poses critical challenges for immunomodulatory therapies that should be accounted for during development and testing of MSCs. In this review, we look to cancer immunotherapy as a model for the challenges MSCs may face in obese environments. We then outline current evidence that obesity alters MSC immunomodulatory function, drastically modifies the host immune system, and therefore reshapes interactions between MSCs and immune cells. Finally, we argue that obese environments may alter essential features of allogeneic MSCs and offer potential strategies for licensing of MSCs to enhance their efficacy in the obese microenvironment. Our aim is to combine insights from basic research in MSC biology and clinical trials to inform new strategies to ensure MSC therapy is effective for a broad range of patients.
Collapse
Affiliation(s)
- Lauren Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Laura Melanie Bitterlich
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
| | - Andrew E. Hogan
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- *Correspondence: James A. Ankrum, ; Karen English,
| | - Karen English
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
- *Correspondence: James A. Ankrum, ; Karen English,
| |
Collapse
|
11
|
Abbaszadeh H, Ghorbani F, Abbaspour-Aghdam S, Kamrani A, Valizadeh H, Nadiri M, Sadeghi A, Shamsasenjan K, Jadidi-Niaragh F, Roshangar L, Ahmadi M. Chronic obstructive pulmonary disease and asthma: mesenchymal stem cells and their extracellular vesicles as potential therapeutic tools. Stem Cell Res Ther 2022; 13:262. [PMID: 35725505 PMCID: PMC9208161 DOI: 10.1186/s13287-022-02938-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/31/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and asthma, are one of the most frequent causes of morbidity and mortality in the global. COPD is characterized by progressive loss of lung function through inflammation, apoptosis, and oxidative stress caused by chronic exposure to harmful environmental pollutants. Airway inflammation and epithelial remodeling are also two main characteristics of asthma. In spite of extensive efforts from researchers, there is still a great need for novel therapeutic approaches for treatment of these conditions. Accumulating evidence suggests the potential role of mesenchymal stem cells (MSCs) in treatment of many lung injuries due to their beneficial features including immunomodulation and tissue regeneration. Besides, the therapeutic advantages of MSCs are chiefly related to their paracrine functions such as releasing extracellular vesicles (EVs). EVs comprising exosomes and microvesicles are heterogeneous bilayer membrane structures loaded with various lipids, nucleic acids and proteins. Due to their lower immunogenicity, tumorigenicity, and easier management, EVs have appeared as favorable alternatives to stem cell therapies. Therefore, in this review, we provided an overview on the current understanding of the importance of MSCs and MSC-derived EVs from different sources reported in preclinical and clinical COPD and asthmatic models.
Collapse
Affiliation(s)
- Hossein Abbaszadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Ghorbani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amin Kamrani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Valizadeh
- Tuberculosis and Lung Disease Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Nadiri
- Tuberculosis and Lung Disease Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Sadeghi
- Tuberculosis and Lung Disease Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Chen R, Hao Z, Wang Y, Zhu H, Hu Y, Chen T, Zhang P, Li J. Mesenchymal Stem Cell-Immune Cell Interaction and Related Modulations for Bone Tissue Engineering. Stem Cells Int 2022; 2022:7153584. [PMID: 35154331 PMCID: PMC8825274 DOI: 10.1155/2022/7153584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Critical bone defects and related delayed union and nonunion are still worldwide problems to be solved. Bone tissue engineering is mainly aimed at achieving satisfactory bone reconstruction. Mesenchymal stem cells (MSCs) are a kind of pluripotent stem cells that can differentiate into bone cells and can be used as one of the key pillars of bone tissue engineering. In recent decades, immune responses play an important role in bone regeneration. Innate immune responses provide a suitable inflammatory microenvironment for bone regeneration and initiate bone regeneration in the early stage of fracture repair. Adaptive immune responses maintain bone regeneration and bone remodeling. MSCs and immune cells regulate each other. All kinds of immune cells and secreted cytokines can regulate the migration, proliferation, and osteogenic differentiation of MSCs, which have a strong immunomodulatory ability to these immune cells. This review mainly introduces the interaction between MSCs and immune cells on bone regeneration and its potential mechanism, and discusses the practical application in bone tissue engineering by modulating this kind of cell-to-cell crosstalk. Thus, an in-depth understanding of these principles of bone immunology can provide a new way for bone tissue engineering.
Collapse
Affiliation(s)
- Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hongzhen Zhu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Department of Orthopedics, Suzhou Science and Technology Town Hospital, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou 215153, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
13
|
Mo Y, Kim Y, Bang JY, Jung J, Lee CG, Elias JA, Kang HR. Mesenchymal Stem Cells Attenuate Asthmatic Inflammation and Airway Remodeling by Modulating Macrophages/Monocytes in the IL-13-Overexpressing Mouse Model. Immune Netw 2022; 22:e40. [DOI: 10.4110/in.2022.22.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yosep Mo
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yujin Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Young Bang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jiung Jung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chun-Geun Lee
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Jack A. Elias
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Pang QM, Chen SY, Fu SP, Zhou H, Zhang Q, Ao J, Luo XP, Zhang T. Regulatory Role of Mesenchymal Stem Cells on Secondary Inflammation in Spinal Cord Injury. J Inflamm Res 2022; 15:573-593. [PMID: 35115806 PMCID: PMC8802142 DOI: 10.2147/jir.s349572] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Hui Zhou
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Jun Ao
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Xiao-Ping Luo
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Correspondence: Tao Zhang; Qian Zhang, Email ;
| |
Collapse
|
15
|
Yang Y, Wang Y. Autocrine, Paracrine, and Endocrine Signals That Can Alter Alveolar Macrophages Function. Rev Physiol Biochem Pharmacol 2022; 186:177-198. [PMID: 36472676 DOI: 10.1007/112_2022_76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar macrophages (AMs) are extremely versatile cells with complex functions involved in health or diseases such as pneumonia, asthma, and pulmonary alveolar proteinosis. In recent years, it has been widely identified that the different functions and states of macrophages are the results from the complex interplay between microenvironmental signals and macrophage lineage. Diverse and complicated signals to which AMs respond are mentioned when they are described individually or in a particular state of AMs. In this review, the microenvironmental signals are divided into autocrine, paracrine, and endocrine signals based on their secreting characteristics. This new perspective on classification provides a more comprehensive and systematic introduction to the complex signals around AMs and is helpful for understanding the roles of AMs affected by physiological environment. The existing possible treatments of AMs are also mentioned in it. The thorough understanding of AMs signals modulation may be contributed to the development of more effective therapies for AMs-related lung diseases.
Collapse
Affiliation(s)
- Yue Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
16
|
Shin JW, Ryu S, Ham J, Jung K, Lee S, Chung DH, Kang HR, Kim HY. Mesenchymal Stem Cells Suppress Severe Asthma by Directly Regulating Th2 Cells and Type 2 Innate Lymphoid Cells. Mol Cells 2021; 44:580-590. [PMID: 34462397 PMCID: PMC8424137 DOI: 10.14348/molcells.2021.0101] [Citation(s) in RCA: 15] [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: 04/19/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022] Open
Abstract
Patients with severe asthma have unmet clinical needs for effective and safe therapies. One possibility may be mesenchymal stem cell (MSC) therapy, which can improve asthma in murine models. However, it remains unclear how MSCs exert their beneficial effects in asthma. Here, we examined the effect of human umbilical cord blood-derived MSCs (hUC-MSC) on two mouse models of severe asthma, namely, Alternaria alternata-induced and house dust mite (HDM)/diesel exhaust particle (DEP)-induced asthma. hUC-MSC treatment attenuated lung type 2 (Th2 and type 2 innate lymphoid cell) inflammation in both models. However, these effects were only observed with particular treatment routes and timings. In vitro co-culture showed that hUC-MSC directly downregulated the interleukin (IL)-5 and IL-13 production of differentiated mouse Th2 cells and peripheral blood mononuclear cells from asthma patients. Thus, these results showed that hUC-MSC treatment can ameliorate asthma by suppressing the asthmogenic cytokine production of effector cells. However, the successful clinical application of MSCs in the future is likely to require careful optimization of the route, dosage, and timing.
Collapse
Affiliation(s)
- Jae Woo Shin
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seungwon Ryu
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jongho Ham
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Keehoon Jung
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
17
|
Moradinasab S, Pourbagheri-Sigaroodi A, Zafari P, Ghaffari SH, Bashash D. Mesenchymal stromal/stem cells (MSCs) and MSC-derived extracellular vesicles in COVID-19-induced ARDS: Mechanisms of action, research progress, challenges, and opportunities. Int Immunopharmacol 2021; 97:107694. [PMID: 33932694 PMCID: PMC8079337 DOI: 10.1016/j.intimp.2021.107694] [Citation(s) in RCA: 20] [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: 03/16/2021] [Revised: 04/11/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023]
Abstract
In late 2019, a novel coronavirus (SARS-CoV-2) emerged in Wuhan city, Hubei province, China. Rapidly escalated into a worldwide pandemic, it has caused an unprecedented and devastating situation on the global public health and society economy. The severity of recent coronavirus disease, abbreviated to COVID-19, seems to be mostly associated with the patients' immune response. In this vein, mesenchymal stromal/stem cells (MSCs) have been suggested as a worth-considering option against COVID-19 as their therapeutic properties are mainly displayed in immunomodulation and anti-inflammatory effects. Indeed, administration of MSCs can attenuate cytokine storm and enhance alveolar fluid clearance, endothelial recovery, and anti-fibrotic regeneration. Despite advantages attributed to MSCs application in lung injuries, there are still several issues __foremost probability of malignant transformation and incidence of MSCs-related coagulopathy__ which should be resolved for the successful application of MSC therapy in COVID-19. In the present study, we review the historical evidence of successful use of MSCs and MSC-derived extracellular vesicles (EVs) in the treatment of acute respiratory distress syndrome (ARDS). We also take a look at MSCs mechanisms of action in the treatment of viral infections, and then through studying both the dark and bright sides of this approach, we provide a thorough discussion if MSC therapy might be a promising therapeutic approach in COVID-19 patients.
Collapse
Affiliation(s)
- Susan Moradinasab
- Iranian Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Zafari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Refaie AF, Elbassiouny BL, Kloc M, Sabek OM, Khater SM, Ismail AM, Mohamed RH, Ghoneim MA. From Mesenchymal Stromal/Stem Cells to Insulin-Producing Cells: Immunological Considerations. Front Immunol 2021; 12:690623. [PMID: 34248981 PMCID: PMC8262452 DOI: 10.3389/fimmu.2021.690623] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy for type 1 diabetes mellitus (T1DM) has been the subject matter of many studies over the past few decades. The wide availability, negligible teratogenic risks and differentiation potential of MSCs promise a therapeutic alternative to traditional exogenous insulin injections or pancreatic transplantation. However, conflicting arguments have been reported regarding the immunological profile of MSCs. While some studies support their immune-privileged, immunomodulatory status and successful use in the treatment of several immune-mediated diseases, others maintain that allogeneic MSCs trigger immune responses, especially following differentiation or in vivo transplantation. In this review, the intricate mechanisms by which MSCs exert their immunomodulatory functions and the influencing variables are critically addressed. Furthermore, proposed avenues to enhance these effects, including cytokine pretreatment, coadministration of mTOR inhibitors, the use of Tregs and gene manipulation, are presented. As an alternative, the selection of high-benefit, low-risk donors based on HLA matching, PD-L1 expression and the absence of donor-specific antibodies (DSAs) are also discussed. Finally, the necessity for the transplantation of human MSC (hMSC)-derived insulin-producing cells (IPCs) into humanized mice is highlighted since this strategy may provide further insights into future clinical applications.
Collapse
Affiliation(s)
- Ayman F Refaie
- Nephrology Department, Urology and Nephrology Center, Mansoura, Egypt
| | | | - Malgorzata Kloc
- Department of Immunobiology, The Houston Methodist Research Institute, Houston, TX, United States.,Department of Surgery, The Houston Methodist Hospital, Houston, TX, United States.,Department of Genetics, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| | - Omaima M Sabek
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, United States.,Department of Cell and Microbiology Biology, Weill Cornell Medical Biology, New York, NY, United States
| | - Sherry M Khater
- Pathology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Amani M Ismail
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Rania H Mohamed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
19
|
Cui Z, Feng Y, Li D, Li T, Gao P, Xu T. Activation of aryl hydrocarbon receptor (AhR) in mesenchymal stem cells modulates macrophage polarization in asthma. J Immunotoxicol 2021; 17:21-30. [PMID: 31922435 DOI: 10.1080/1547691x.2019.1706671] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Macrophage polarization has been demonstrated to exert a vital role on asthma pathogenesis. Mesenchymal stem cells (MSC) have the capacity to modulate macrophage differentiation from a pro-inflammatory M1 phenotype toward an anti-inflammatory M2 phenotype. However, the impact of MSC-macrophage interactions on asthma development and underlying mechanisms responsible for this interaction remain largely unknown. The aim of this study was to investigate the role of AhR expressed on MSC in macrophage polarization in a cockroach extract (CRE)-induced asthma mouse model. The studies here revealed that MSC polarized macrophages from a pro-inflammatory M1 phenotype toward an anti-inflammatory M2 phenotype in this model. The mRNA levels of interleukin (IL)-6, IL-1β, and NOS2 as M1 markers were significantly decreased while those of select M2 markers such as Arg-1, FIZZ1, and YM-1 were significantly enhanced. It was also observed that aryl hydrocarbon receptor (AhR) signaling was significantly increased during asthma pathogenesis as demonstrated by enhanced mRNA expression of AhR, CYP1a1, and CYP1b1. It was also seen that the elevated AhR signaling was able to attenuate the onset of asthma. Use of an AhR antagonist (CH223191) resulted in significant inhibition of the AhR signaling and increases in M2 marker expression, but led to elevation of expression of M1 markers in the CRE-induced asthma model. Taken together, the current study showed that MSC can modulate macrophage polarization, in part, via activation of AhR signaling during CRE-induced asthma.
Collapse
Affiliation(s)
- Zhuang Cui
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Feng
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danqing Li
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Taoping Li
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ting Xu
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Guo H, Su Y, Deng F. Effects of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Lung Diseases: Current Status and Future Perspectives. Stem Cell Rev Rep 2021; 17:440-458. [PMID: 33211245 PMCID: PMC7675022 DOI: 10.1007/s12015-020-10085-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal cells (MSCs) as a kind of pluripotent adult stem cell have shown great therapeutic potential in relation to many diseases in anti-inflammation and regeneration. The results of preclinical experiments and clinical trials have demonstrated that MSC-derived secretome possesses immunoregulatory and reparative abilities and that this secretome is capable of modulating innate and adaptive immunity and reprograming the metabolism of recipient cells via paracrine mechanisms. It has been recognized that MSC-derived secretome, including soluble proteins (cytokines, chemokines, growth factors, proteases), extracellular vesicles (EVs) and organelles, plays a key role in tissue repair and regeneration in bronchopulmonary dysplasia, acute respiratory distress syndrome (ARDS), bronchial asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension, and silicosis. This review summarizes the known functions of MSC-EV modulation in lung diseases, coupled with the future challenges of MSC-EVs as a new pharmaceutical agent. The identification of underlying mechanisms for MSC-EV might provide a new direction for MSC-centered treatment in lung diseases.Graphical abstract.
Collapse
Affiliation(s)
- Haiyan Guo
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, 230022 Hefei, Anhui Province People’s Republic of China
| | - Yue Su
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast, Belfast, BT9 7BL UK
| | - Fang Deng
- Department of Nephrology, Anhui Provincial Children’s Hospital, Hefei City, Anhui Province 230022 People’s Republic of China
| |
Collapse
|
21
|
Tynecka M, Moniuszko M, Eljaszewicz A. Old Friends with Unexploited Perspectives: Current Advances in Mesenchymal Stem Cell-Based Therapies in Asthma. Stem Cell Rev Rep 2021; 17:1323-1342. [PMID: 33649900 PMCID: PMC7919631 DOI: 10.1007/s12015-021-10137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have a great regenerative and immunomodulatory potential that was successfully tested in numerous pre-clinical and clinical studies of various degenerative, hematological and inflammatory disorders. Over the last few decades, substantial immunoregulatory effects of MSC treatment were widely observed in different experimental models of asthma. Therefore, it is tempting to speculate that stem cell-based treatment could become an attractive means to better suppress asthmatic airway inflammation, especially in subjects resistant to currently available anti-inflammatory therapies. In this review, we discuss mechanisms accounting for potent immunosuppressive properties of MSCs and the rationale for their use in asthma. We describe in detail an intriguing interplay between MSCs and other crucial players in the immune system as well as lung microenvironment. Finally, we reveal the potential of MSCs in maintaining airway epithelial integrity and alleviating lung remodeling.
Collapse
Affiliation(s)
- Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
- Department of Allergology and Internal Medicine, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, Białystok, 15-276, Poland.
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
| |
Collapse
|
22
|
Ren J, Liu Y, Yao Y, Feng L, Zhao X, Li Z, Yang L. Intranasal delivery of MSC-derived exosomes attenuates allergic asthma via expanding IL-10 producing lung interstitial macrophages in mice. Int Immunopharmacol 2021; 91:107288. [PMID: 33360827 DOI: 10.1016/j.intimp.2020.107288] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) have been investigated in preventing and treating allergic asthma in many reports. Recently, MSC-derived exosomes (MSC-Exo) were showed a promising alternative to stem cell-based therapy in many kinds of diseases. However, the effect of MSC-Exo on allergic asthma has not been investigated thoroughly thus far. Here, we aimed to investigate the immunomodulation effect of MSC-Exo in a murine model of asthma and explore the underlying mechanisms. BALB/c mice were sensitized and challenged by OVA to establish asthma model. MSC-Exo were intranasally delivered before or during challenge and the protective effect were assessed after the last OVA challenge. Allergic airway inflammation elicited by OVA were significantly attenuated by intranasal delivery of MSC-Exo. To explore the protective mechanism of MSC-Exo, lung interstitial macrophages (IMs) and alveolar macrophages (AMs) were analyzed by flow cytometry and the origin of IMs were traced. Lung IMs ratios were significantly enhanced and high level of IL-10 was produced after MSC-Exo intranasal delivery. IMs ratios were not obviously affected by CCR2 inhibitor or Clodronate liposome administration, whereas significantly decreased in splenectomized mice. Cx3cr1+ cell specific IL-10 conditionally deficient mice were used to further examine the role of IL-10 producing IMs in allergic asthma. IMs-mediated protection was dependent on IL-10, given that the protection disappeared in Cx3cr1-IL-10-/-mice. In conclusion, intranasal delivery of MSC-Exo could substantially expand lung IL-10-producing IMs, which may originate from spleen, thus contribute to protection against allergic asthma in mice.
Collapse
Affiliation(s)
- Jiling Ren
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Yongzhe Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ye Yao
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Lifeng Feng
- School of Medicine, Nankai University, Tianjin, China
| | - Xiaotong Zhao
- School of Medicine, Nankai University, Tianjin, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, China.
| | - Liang Yang
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
23
|
Ridzuan N, Zakaria N, Widera D, Sheard J, Morimoto M, Kiyokawa H, Mohd Isa SA, Chatar Singh GK, Then KY, Ooi GC, Yahaya BH. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD). Stem Cell Res Ther 2021; 12:54. [PMID: 33436065 PMCID: PMC7805108 DOI: 10.1186/s13287-020-02088-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an incurable and debilitating chronic disease characterized by progressive airflow limitation associated with abnormal levels of tissue inflammation. Therefore, stem cell-based approaches to tackle the condition are currently a focus of regenerative therapies for COPD. Extracellular vesicles (EVs) released by all cell types are crucially involved in paracrine, extracellular communication. Recent advances in the field suggest that stem cell-derived EVs possess a therapeutic potential which is comparable to the cells of their origin. METHODS In this study, we assessed the potential anti-inflammatory effects of human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs in a rat model of COPD. EVs were isolated from hUC-MSCs and characterized by the transmission electron microscope, western blotting, and nanoparticle tracking analysis. As a model of COPD, male Sprague-Dawley rats were exposed to cigarette smoke for up to 12 weeks, followed by transplantation of hUC-MSCs or application of hUC-MSC-derived EVs. Lung tissue was subjected to histological analysis using haematoxylin and eosin staining, Alcian blue-periodic acid-Schiff (AB-PAS) staining, and immunofluorescence staining. Gene expression in the lung tissue was assessed using microarray analysis. Statistical analyses were performed using GraphPad Prism 7 version 7.0 (GraphPad Software, USA). Student's t test was used to compare between 2 groups. Comparison among more than 2 groups was done using one-way analysis of variance (ANOVA). Data presented as median ± standard deviation (SD). RESULTS Both transplantation of hUC-MSCs and application of EVs resulted in a reduction of peribronchial and perivascular inflammation, alveolar septal thickening associated with mononuclear inflammation, and a decreased number of goblet cells. Moreover, hUC-MSCs and EVs ameliorated the loss of alveolar septa in the emphysematous lung of COPD rats and reduced the levels of NF-κB subunit p65 in the tissue. Subsequent microarray analysis revealed that both hUC-MSCs and EVs significantly regulate multiple pathways known to be associated with COPD. CONCLUSIONS In conclusion, we show that hUC-MSC-derived EVs effectively ameliorate by COPD-induced inflammation. Thus, EVs could serve as a new cell-free-based therapy for the treatment of COPD.
Collapse
Affiliation(s)
- Noridzzaida Ridzuan
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Jonathan Sheard
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Mitsuru Morimoto
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Hirofumi Kiyokawa
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Seoparjoo Azmel Mohd Isa
- Department of Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Malaysia
| | - Gurjeet Kaur Chatar Singh
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Kong-Yong Then
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Ghee-Chien Ooi
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia.
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| |
Collapse
|
24
|
Lin S, Lin M, Ma H, Wang X, Zhang D, Wu W, Lin J, Gao H. Identification of miR-4793-3p as a potential biomarker for bacterial infection in patients with hepatitis B virus-related liver cirrhosis: A pilot study. Exp Ther Med 2020; 21:120. [PMID: 33335583 PMCID: PMC7739867 DOI: 10.3892/etm.2020.9552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus-related liver cirrhosis (HBV-LC) is susceptible to bacterial infections, which could lead to adverse prognosis in patients. MicroRNAs (miRs/miRNAs) are easily detected in peripheral blood and are involved in multiple liver diseases. The present pilot study aimed to investigate differentially expressed (DE) miRNAs in the serum of patients with HBV-LC and bacterial infection, and to identify potential biomarkers. The first batch of clinical samples was collected, including four patients with HBV-LC and infection, four patients with HBV-LC without infection, four patients with chronic hepatitis B (CHB) and four healthy controls. miRNA expression was analyzed by Affymetrix GeneChip miRNA 4.0 Array. A total of 385 DE miRNAs (upregulated, 160; downregulated, 225) were detected in patients with HBV-LC and infection compared with patients with HBV-LC without infection. miR-4793-3p was significantly upregulated in patients with HBV-LC and infection compared with its levels in the other three groups: HBV-LC without infection [log-transformed fold change (logFC)=7.96; P=0.0458), CHB (logFC=34.53; P=0.0003) and healthy controls (logFC=3.34; P=0.0219)]. Reverse transcription-quantitative PCR (RT-qPCR) was performed to validate miR-4793-3p expression in another batch of clinical samples. RT-qPCR showed that miR-4793-3p was highly expressed in patients with HBV-LC and infection compared with its levels in patients with HBV-LC without infection (P<0.05). The non-parametric random forest regression model was built to access the diagnostic value of miR-4793-3p, and the receiver operating characteristic curve demonstrated that the area under the curve was 92.2%. Target gene analysis with bioinformatics tools and Gene Expression Omnibus data (GSE46955) showed that miR-4793-3p could participate in the TGF-β signaling pathway. Functional experiments revealed that overexpressed miR-4793-3p could impair TGF-β function by downregulating Gremlin-1. The present pilot study suggests that miR-4793-3p could be a feasible indicator for bacterial infection in patients with HBV-LC, and it would be valuable for further research.
Collapse
Affiliation(s)
- Shenglong Lin
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Minghua Lin
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Huaxi Ma
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Xiangmei Wang
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Dongqing Zhang
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Wenjun Wu
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Jiahuang Lin
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Haibing Gao
- Department of Severe Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Fuzhou Infectious Diseases Hospital, Fuzhou, Fujian 350002, P.R. China.,Department of Hepatology, Infectious Diseases Hospital of Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| |
Collapse
|
25
|
Yu X, Yu L, Guo B, Chen R, Qiu C. A narrative review of research advances in mesenchymal stem cell therapy for asthma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1461. [PMID: 33313206 PMCID: PMC7723541 DOI: 10.21037/atm-20-6389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Asthma is a chronic inflammatory disease of the airways that involves multiple cells, including inflammatory cells, structural cells, and cellular components. Glucocorticoids and beta-receptor agonists are still the first choices for asthma treatment. However, the asthma symptoms may still be poorly controlled in some patients after an optimal treatment. Mesenchymal stem cells (MSCs) are characterized by the potential for multi-directional differentiation and can exert immunomodulatory and anti-inflammatory effects. Its role in treating asthma has increasingly been recognized in recent years. In this review article, we sought to summarize the recent advances in the therapeutic effects of MSCs on several types of asthma and explain the relevant mechanisms. Articles on asthma treatment with MSCs as of January 2020 were searched in PubMed, Google Scholar, and Web of Science databases. It was found that MSCs have therapeutic effects on allergic asthma, non-allergic asthma and occupational asthma; gene-modified or pretreated MSCs improves the therapeutic effects of MSCs in asthma; MSC-derived conditioned medium or extracellular vesicles possess the considerable curative effect as MSC on asthma; and MSCs exert their therapeutic effects on asthma by restoring Th1/Th2 balance, reversing Th17/Tregs imbalance, inhibiting DC maturation, and promoting the switch of M1 to M2 and repairing epithelial injury. Thus, MSCs may be a promising treatment for asthma.
Collapse
Affiliation(s)
- Xiu Yu
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital (Second Clinical Medical College of Jinan University & First Affiliated Hospital of Southern University of Science and Technology), Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Li Yu
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital (Second Clinical Medical College of Jinan University & First Affiliated Hospital of Southern University of Science and Technology), Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Bingxin Guo
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital (Second Clinical Medical College of Jinan University & First Affiliated Hospital of Southern University of Science and Technology), Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Rongchang Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital (Second Clinical Medical College of Jinan University & First Affiliated Hospital of Southern University of Science and Technology), Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital (Second Clinical Medical College of Jinan University & First Affiliated Hospital of Southern University of Science and Technology), Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| |
Collapse
|
26
|
Jorde I, Hildebrand CB, Kershaw O, Lücke E, Stegemann-Koniszewski S, Schreiber J. Modulation of Allergic Sensitization and Allergic Inflammation by Staphylococcus aureus Enterotoxin B in an Ovalbumin Mouse Model. Front Immunol 2020; 11:592186. [PMID: 33193436 PMCID: PMC7649385 DOI: 10.3389/fimmu.2020.592186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023] Open
Abstract
The superantigen Staphylococcus aureus (S. aureus) enterotoxin B (SEB) has been proposed a central player in the associations between S. aureus nasal colonization and the development of allergic asthma. Previously, SEB has been shown to aggravate allergic sensitization and allergic airway inflammation (AAI) in experimental mouse models. Aiming at understanding the underlying immunological mechanisms, we tested the hypothesis that intranasal (i.n.) SEB-treatment divergently modulates AAI depending on the timing and intensity of the SEB-encounter. In an ovalbumin-mediated mouse model of AAI, we treated mice i.n. with 50 ng or 500 ng SEB either together with the allergic challenge or prior to the peripheral sensitization. We observed SEB to affect different hallmark parameters of AAI depending on the timing and the dose of treatment. SEB administered i.n. together with the allergic challenge significantly modulated respiratory leukocyte accumulation, intensified lymphocyte activation and, at the higher dose, induced a strong type-1 and pro-inflammatory cytokine response and alleviated airway hyperreactivity in AAI. SEB administered i.n. prior to the allergic sensitization at the lower dose significantly boosted the specific IgE response while administration of the higher dose led to a significantly reduced recruitment of immune cells, including eosinophils, to the respiratory tract and to a significantly dampened Th-2 cytokine response without inducing a Th-1 or pro-inflammatory response. We show a remarkably versatile potential for SEB to either aggravate or alleviate different parameters of allergic sensitization and AAI. Our study thereby not only highlights the complexity of the associations between S. aureus and allergic asthma but possibly even points at prophylactic and therapeutic pathways.
Collapse
Affiliation(s)
- Ilka Jorde
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Christina B Hildebrand
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Olivia Kershaw
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Eva Lücke
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
27
|
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: 5.3] [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
|
28
|
Jaiswal A, Maurya M, Maurya P, Barthwal MK. Lin28B Regulates Angiotensin II-Mediated Let-7c/miR-99a MicroRNA Formation Consequently Affecting Macrophage Polarization and Allergic Inflammation. Inflammation 2020; 43:1846-1861. [DOI: 10.1007/s10753-020-01258-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
29
|
Li H, Tian Y, Xie L, Liu X, Huang Z, Su W. Mesenchymal stem cells in allergic diseases: Current status. Allergol Int 2020; 69:35-45. [PMID: 31445840 DOI: 10.1016/j.alit.2019.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Allergic diseases, which include asthma, allergic skin diseases, allergic rhinitis and allergic conjunctivitis, have already garnered worldwide public health attention over recent decades. Mesenchymal stem cells (MSCs) have gradually emerged as a potential method for treating allergic diseases due to their immunosuppressive characteristics, tissue repair ability and secretion of various biological factors. This potential of MSC-based therapy has been confirmed in clinical and preclinical studies, which report the therapeutic benefits of MSCs for various allergic diseases and explore the antiallergic mechanisms. In this review, we focus on the discoveries and biological mechanisms of MSCs as a therapeutic tool in allergic diseases. We discuss the challenges of conducting MSC studies as well as future directions.
Collapse
|
30
|
Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif 2019; 53:e12712. [PMID: 31730279 PMCID: PMC6985662 DOI: 10.1111/cpr.12712] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/11/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be derived from various adult tissues with multipotent and self‐renewal abilities. The characteristics of presenting no major ethical concerns, having low immunogenicity and possessing immune modulation functions make MSCs promising candidates for stem cell therapies. MSCs could promote inflammation when the immune system is underactivated and restrain inflammation when the immune system is overactivated to avoid self‐overattack. These cells express many immune suppressors to switch them from a pro‐inflammatory phenotype to an anti‐inflammatory phenotype, resulting in immune effector cell suppression and immune suppressor cell activation. We would discuss the mechanisms governing the immune modulation function of these cells in this review, especially the immune‐suppressive effects of MSCs.
Collapse
Affiliation(s)
- Wei Jiang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
31
|
Curtis BJ, Shults JA, Boe DM, Ramirez L, Kovacs EJ. Mesenchymal stem cell treatment attenuates liver and lung inflammation after ethanol intoxication and burn injury. Alcohol 2019; 80:139-148. [PMID: 30217504 DOI: 10.1016/j.alcohol.2018.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
Cutaneous burn injury is one of the most devastating injuries one can obtain, with tissue damage extending beyond the skin wound to distal organs, including the gastrointestinal tract, liver, and lungs. Multiple organ failure is a leading cause of death after burn injury, resulting in excessive systemic and localized inflammation directly contributing to end organ damage. We postulated that the gut-liver-lung inflammatory axis underscores multiple organ failure in the context of burn injury and is hyper-activated when ethanol intoxication precedes burn. Mesenchymal stem cells (MSCs) are regenerative and anti-inflammatory, and MSC treatment has been shown to be beneficial in several immune disorders and injury models. Our objective was to determine whether intravenous infusion of exogenous bone marrow-derived MSCs could reduce post-burn and intoxication pulmonary, hepatic, and systemic inflammation. Vehicle- or ethanol- (1.6 g/kg) treated mice were subjected to sham or 15% total body surface area scald burn. One hour post-injury, mice were given 5 × 105 CFSE-labeled MSCs or phosphate-buffered saline intravenously (i.v.) and were euthanized 24 h later. We assessed circulating biomarkers of inflammation and liver damage, measured cytokine and chemokine production, and quantified apoptosis in lung and liver tissue. Compared to intoxicated and burned mice, those treated with MSCs had less cellularity, limited apoptosis, and a slight reduction in the pro-inflammatory cytokine interleukin-6 (IL-6) and the neutrophil chemokine, KC (CXCL1) in lung tissue. Mice with MSCs treatment had more dramatic anti-inflammatory effects on systemic and hepatic inflammation, as serum IL-6 levels were diminished by 43%, and il6 and kc expression in liver tissue were markedly reduced, as were biomarkers of liver damage, aspartate transaminase (AST) and alanine transaminase (AST), compared with intoxicated and burned mice. Taken together, our results suggest intravenous MSCs treatment can diminish systemic inflammation, lessen hepatic damage, and decrease liver and lung apoptosis and inflammation, indicating MSCs as a novel therapy for restoring homeostasis of multiple organ systems in intoxicated burn patients.
Collapse
Affiliation(s)
- Brenda J Curtis
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Jill A Shults
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, IL, United States
| | - Devin M Boe
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Luis Ramirez
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, IL, United States
| | - Elizabeth J Kovacs
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
32
|
Capcha JMC, Rodrigues CE, Moreira RDS, Silveira MD, Dourado P, Dos Santos F, Irigoyen MC, Jensen L, Garnica MR, Noronha IL, Andrade L, Gomes SA. Wharton's jelly-derived mesenchymal stem cells attenuate sepsis-induced organ injury partially via cholinergic anti-inflammatory pathway activation. Am J Physiol Regul Integr Comp Physiol 2019; 318:R135-R147. [PMID: 31596111 DOI: 10.1152/ajpregu.00098.2018] [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: 12/21/2022]
Abstract
Sepsis induces organ dysfunction due to overexpression of the inflammatory host response, resulting in cardiopulmonary and autonomic dysfunction, thus increasing the associated morbidity and mortality. Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) express genes and secrete factors with anti-inflammatory properties, neurological and immunological protection, as well as improve survival in experimental sepsis. The cholinergic anti-inflammatory pathway (CAP) is mediated by α7-nicotinic acetylcholine receptors (α7nAChRs), which play an important role in the control of systemic inflammation. We hypothesized that WJ-MSCs attenuate sepsis-induced organ injury in the presence of an activated CAP pathway. To confirm our hypothesis, we evaluated the effects of WJ-MSCs as a treatment for cardiopulmonary injury and on neuroimmunomodulation. Male Wistar rats were randomly divided into four groups: control (sham-operated); cecal ligation and puncture (CLP) alone; CLP+WJ-MSCs (1 × 106 cells, at 6 h post-CLP); and CLP+methyllycaconitine (MLA)+WJ-MSCs (5 mg/kg body wt, at 5.5 h post-CLP, and 1 × 106 cells, at 6 h post-CLP, respectively). All experiments, including the assessment of echocardiographic parameters and heart rate variability, were performed 24 h after CLP. WJ-MSC treatment attenuated diastolic dysfunction and restored baroreflex sensitivity. WJ-MSCs also increased cardiac sympathetic and cardiovagal activity. WJ-MSCs reduced leukocyte infiltration and proinflammatory cytokines, effects that were abolished by administration of a selective α7nAChR antagonist (MLA). In addition, WJ-MSC treatment also diminished apoptosis in the lungs and spleen. In cardiac and splenic tissue, WJ-MSCs downregulated α7nAChR expression, as well as reduced the phospho-STAT3-to-total STAT3 ratio in the spleen. WJ-MSCs appear to protect against sepsis-induced organ injury by reducing systemic inflammation, at least in part, via a mechanism that is dependent on an activated CAP.
Collapse
Affiliation(s)
- José Manuel Cóndor Capcha
- Laboratory of Genetics, Cellular Biology, and Molecular Biology, University of São Paulo School of Medicine, São Paulo, Brazil.,Laboratory of Basic Research, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Roberto de Souza Moreira
- Laboratory of Basic Research, University of São Paulo School of Medicine, São Paulo, Brazil.,Federal University of Goiás at Catalão, Catalão, Brazil
| | | | - Paulo Dourado
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Fernando Dos Santos
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Leonardo Jensen
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Margoth Ramos Garnica
- Laboratory of Genetics, Cellular Biology, and Molecular Biology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Irene L Noronha
- Laboratory of Genetics, Cellular Biology, and Molecular Biology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Lúcia Andrade
- Laboratory of Basic Research, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Samirah Abreu Gomes
- Laboratory of Genetics, Cellular Biology, and Molecular Biology, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
33
|
Lin SC, Liou YM, Ling TY, Chuang YH, Chiang BL. Placenta-Derived Mesenchymal Stem Cells Reduce the Interleukin-5 Level Experimentally in Children with Asthma. Int J Med Sci 2019; 16:1430-1438. [PMID: 31673233 PMCID: PMC6818200 DOI: 10.7150/ijms.33590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/21/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) have been investigated as a new treatment option for various diseases in recent years. However, the role of placenta-derived MSCs in children with asthma remains unclear. We assessed the effect of placenta-derived MSCs on T cell immune responses and cytokine IL-5 levels according to cultures in children with and without asthma. Study design: We enrolled children with and without asthma and recorded asthma symptom scores in the asthma group. Blood samples from children were collected to isolate peripheral blood mononuclear cells (PBMCs) and determine the total IgE level. The PBMCs were cultured in vitro with or without MSCs after stimulation with human anti-CD3 and anti-CD28 antibodies (0.5 μg/mL) to evaluate the effect of placenta-derived MSCs. Flow cytometry was performed to detect the activation and proliferation of CD4+ and CD8+ T cells. Pre- and post-culture IL-5 levels were measured in all samples. Results: The percentages of activation and proliferation among CD4+ and CD8+ T cells after coculture with MSCs were significantly lower in the asthma group (P < 0.05). IL-5 levels differed significantly between the PBMC culture and PBMC + MSC (P+S) coculture in the asthma group (P < 0.05). IL-5 levels differed significantly between the PBMC culture and P+S coculture in both the lower (P < 0.05) and higher (P < 0.0005) IgE asthma subgroups. IL-5 levels were also decreased in children with all severities of asthma (P < 0.05). Conclusions: Placenta-derived MSCs exerted an anti-IL-5 effect and reduced the IL-5 level in culture in different subgroups of children with asthma.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yih-Mei Liou
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Hui Chuang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Seo YS, Kim HS, Lee AY, Chun JM, Kim SB, Moon BC, Kwon BI. Codonopsis lanceolata attenuates allergic lung inflammation by inhibiting Th2 cell activation and augmenting mitochondrial ROS dismutase (SOD2) expression. Sci Rep 2019; 9:2312. [PMID: 30783201 PMCID: PMC6381190 DOI: 10.1038/s41598-019-38782-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/24/2018] [Indexed: 01/14/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease induced by the inhalation of allergens, which trigger the activation of T helper type 2 (Th2) cells that release Th2 cytokines. Recently, herbal medicines are being considered a major source of novel agents to treat various diseases. In the present study, we evaluated the anti-asthmatic effects of a Codonopsis lanceolata extract (CLE) and the mechanisms involved in its anti-inflammatory effects. Treatment with CLE reduced infiltration of inflammatory cells, especially eosinophils, and the production of mucus in lung tissues. Levels of Th2 cytokines, such as IL-4, IL-5, and IL-13, and chemokines were also decreased following treatment with CLE. Moreover, Th2 cell proportion in vivo and differentiation in vitro were reduced as evidenced by the decreased expression of GATA3+. Furthermore, the expression of superoxide dismutase (SOD)2, a mitochondrial ROS (mROS) scavenger, was increased, which was related to Th2 cell regulation. Interestingly, treatment with CLE increased the number of macrophages in the lungs and enhanced the immune-suppressive property of macrophages. Our findings indicate that CLE has potential as a novel therapeutic agent to inhibit Th2 cell differentiation by regulating mROS scavenging.
Collapse
Affiliation(s)
- Yun-Soo Seo
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Hyo Seon Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - A Yeong Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Jin Mi Chun
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Sung Bae Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Bo-In Kwon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea. .,Department of Pathology, College of Korean Medicine, Sangji University, Wonju-si, Gangwon-do, 26339, Republic of Korea.
| |
Collapse
|
35
|
Zhang LB, He M. Effect of mesenchymal stromal (stem) cell (MSC) transplantation in asthmatic animal models: A systematic review and meta-analysis. Pulm Pharmacol Ther 2018; 54:39-52. [PMID: 30496803 DOI: 10.1016/j.pupt.2018.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/17/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Over the years, mesenchymal stromal (stem) cells (MSCs) have been pre-clinically applied in the treatment of variety kinds of diseases including asthma and chronic lung diseases. Aim of the current study was to systematically review and to conduct meta-analysis on the published studies of MSC treatment in asthma animal models. METHODS Publications on the MSC and asthma treatment was thoroughly searched in the electronic databases. Statistical analysis was then performed using the Comprehensive Meta-Analysis software (Version 3). Effect of MSC therapy on asthma model was assessed by Hedges's g with 95% confidence intervals (95% CIs). Random effect model was used due to the heterogeneity between the studies. RESULTS Meta-analysis of the 32 included studies showed that MSC transplantation was significantly in favor of attenuating lung injury and remodeling (Hedges's g = -9.104 ± 0.951 with 95% CI: -10.969 ∼ -7.240, P < 0.001) and airway inflammation (Hedges's g = -4.146 ± 0.688 with 95% CI: -5.495 ∼ -2.797, P < 0.001). The mechanism of MSC therapy in asthma seems to be regulating the balance of Th1 cytokine and Th2 cytokines (IFN-γ: Hedges's g = 4.779 ± 1.408 with 95% CI: 1.099-2.725, P < 0.001; IL-4: Hedges's g = -10.781 ± 1.062 with 95% CI: -12.863 ∼ -8.699, P < 0.001; IL-5: Hedges's g = -10.537 ± 1.269 with 95% CI: -13.025 ∼ -8.050, P < 0.001; IL-13: Hedges's g = -6.773 ± 0.788 with 95% CI: -8.318 ∼ -5.229, P < 0.001). CONCLUSION Findings of the current systemic review suggested a potential role for MSCs in asthma treatment although it is still challenging in clinical practice. The mechanisms of MSCs in pre-clinical asthma treatment may be associated with attenuating airway inflammation through regulating Th1 and Th2 cytokines.
Collapse
Affiliation(s)
- Li-Bo Zhang
- Department of Respiratory Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min He
- Department of Respiratory Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
36
|
Abreu SC, Xisto DG, de Oliveira TB, Blanco NG, de Castro LL, Kitoko JZ, Olsen PC, Lopes-Pacheco M, Morales MM, Weiss DJ, Rocco PRM. Serum from Asthmatic Mice Potentiates the Therapeutic Effects of Mesenchymal Stromal Cells in Experimental Allergic Asthma. Stem Cells Transl Med 2018; 8:301-312. [PMID: 30426724 PMCID: PMC6392406 DOI: 10.1002/sctm.18-0056] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/30/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory disease characterized by airway inflammation and remodeling, which can lead to progressive decline of lung function. Although mesenchymal stromal cells (MSCs) have shown beneficial immunomodulatory properties in preclinical models of allergic asthma, effects on airway remodeling have been limited. Mounting evidence suggests that prior exposure of MSCs to specific inflammatory stimuli or environments can enhance their immunomodulatory properties. Therefore, we investigated whether stimulating MSCs with bronchoalveolar lavage fluid (BALF) or serum from asthmatic mice could potentiate their therapeutic properties in experimental asthma. In a house dust mite (HDM) extract asthma model in mice, unstimulated, asthmatic BALF‐stimulated, or asthmatic serum‐stimulated MSCs were administered intratracheally 24 hours after the final HDM challenge. Lung mechanics and histology; BALF protein, cellularity, and biomarker levels; and lymph‐node and bone marrow cellularity were assessed. Compared with unstimulated or BALF‐stimulated MSCs, serum‐stimulated MSCs further reduced BALF levels of interleukin (IL)‐4, IL‐13, and eotaxin, total and differential cellularity in BALF, bone marrow and lymph nodes, and collagen fiber content, while increasing BALF IL‐10 levels and improving lung function. Serum stimulation led to higher MSC apoptosis, expression of various mediators (transforming growth factor‐β, interferon‐γ, IL‐10, tumor necrosis factor‐α‐stimulated gene 6 protein, indoleamine 2,3‐dioxygenase‐1, and IL‐1 receptor antagonist), and polarization of macrophages to M2 phenotype. In conclusion, asthmatic serum may be a novel strategy to potentiate therapeutic effects of MSCs in experimental asthma, leading to further reductions in both inflammation and remodeling than can be achieved with unstimulated MSCs. stem cells translational medicine2019;8:301&312
Collapse
Affiliation(s)
- Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tainá B de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia G Blanco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lígia Lins de Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jamil Zola Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, College of Medicine, Burlington, Vermont, USA
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
37
|
de Araújo Farias V, Carrillo-Gálvez AB, Martín F, Anderson P. TGF-β and mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer. Cytokine Growth Factor Rev 2018; 43:25-37. [PMID: 29954665 DOI: 10.1016/j.cytogfr.2018.06.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/30/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) represent a promising cell-based therapy in regenerative medicine and for the treatment of inflammatory/autoimmune diseases. Importantly, MSCs have emerged as an important contributor to the tumor stroma with both pro- and anti-tumorigenic effects. However, the successful translation of MSCs to the clinic and the prevention of their tumorigenic and metastatic effect require a greater understanding of factors controlling their proliferation, differentiation, migration and immunomodulation in vitro and in vivo. The transforming growth factor(TGF)-β1, 2 and 3 are involved in almost every aspect of MSC function. The aim of this review is to highlight the roles that TGF-β play in the biology and therapeutic applications of MSCs. We will discuss the how TGF-β modulate MSC function as well as the paracrine effects of MSC-derived TGF-β on other cell types in the context of tissue regeneration, immune responses and cancer. Finally, taking all these aspects into consideration we discuss how modulation of TGF-β signaling/production in MSCs could be of clinical interest.
Collapse
Affiliation(s)
- Virgínea de Araújo Farias
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; Facultad de Odontología, Universidad de Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - Ana Belén Carrillo-Gálvez
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Francisco Martín
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Per Anderson
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain.
| |
Collapse
|
38
|
Yu Y, Wang D, Li H, Liu Y, Xiang Z, Wu J, Jing X. IPSC‑MSC inhibition assessment in Raw 264.7 cells following oxygen and glucose deprivation reveals a distinct function for cardiopulmonary resuscitation. Mol Med Rep 2018; 17:8212-8220. [PMID: 29658608 PMCID: PMC5983996 DOI: 10.3892/mmr.2018.8864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023] Open
Abstract
Hypoxia is a serious stress state. The nervous system is less tolerant to hypoxia, and cell death due to hypoxia is irreversible. With the incidence of cardiovascular disease gradually increasing, the sudden cardiac death rate is additionally increasing. Although cardiopulmonary resuscitation (CPR) is an important development, recovery is frequently poor. In a successful recovery population, ~40% of the population was in a vegetative state or subsequently succumbed to their condition, and ~20% had brain damage. Therefore, the recovery of the brain is of particular importance in CPR. Immune disorders are one of the major mechanisms of cerebral resuscitation following CPR. Studies have demonstrated that induced pluripotent stem cell-derived mesenchymal stem cells (IPSC-MSCs) have a strong immune regulatory effect during tissue repair and anti-inflammatory effects. IPSC-MSCs may inhibit the inflammatory response by means of the inflammatory reaction network to improve brain function following CPR, although the cellular and molecular mechanisms remain unclear. Macrophages are a bridge between innate immune and specific immune responses in the body; therefore, it was hypothesized that macrophages may be the important effector cell of the role of IPSC-MSCs in improving brain function following recovery of spontaneous respiration and circulation subsequent to cardiopulmonary resuscitation. In the present study, IPSC-MSCs were applied to the oxygen and glucose deprivation (OGD) model. It was observed that intervention with IPSC-MSCs was able to alter the polarization direction of macrophages. The difference in the proportions of M1 and M2 macrophages was statistically significant at 6, 12, 24 and 48 h (P=0.037, P<0.05) in the OGD + IPSC-MSCs group (M1, 33.48±5.6%; M2, 50.84±6.9%) and in the OGD group (M1, 83.55±7.3%; M2, 11.41±3.2%), and over time this trend was more obvious. The polarization direction of macrophages is associated with the neurogenic locus notch homolog protein 1 (Notch-1) signaling pathway. In conclusion, it was observed that IPSC-MSCs may be associated with altered macrophage polarization, which may be accomplished by inhibiting the Notch-1 signaling pathway.
Collapse
Affiliation(s)
- Yi Yu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Dongping Wang
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Li
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yujie Liu
- Department of Breast Surgery and Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat‑Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhao Xiang
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junlin Wu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoli Jing
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
39
|
Takeda K, Webb TL, Ning F, Shiraishi Y, Regan DP, Chow L, Smith MJ, Ashino S, Guth AM, Hopkins S, Gelfand EW, Dow S. Mesenchymal Stem Cells Recruit CCR2 + Monocytes To Suppress Allergic Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 200:1261-1269. [PMID: 29352000 DOI: 10.4049/jimmunol.1700562] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 12/03/2017] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSC) exert immune modulatory properties and previous studies demonstrated suppressive effects of MSC treatment in animal models of allergic airway inflammation. However, the underlying mechanisms have not been fully elucidated. We studied the role of MSC in immune activation and subsequent recruitment of monocytes in suppressing airway hyperresponsiveness and airway inflammation using a mouse model of allergic airway inflammation. MSC administration prior to or after allergen challenge inhibited the development of airway inflammation in allergen-sensitized mice. This was accompanied by an influx of CCR2-positive monocytes, which were localized around injected MSC in the lungs. Notably, IL-10-producing monocytes and/or macrophages were also increased in the lungs. Systemic administration of liposomal clodronate or a CCR2 antagonist significantly prevented the suppressive effects of MSC. Activation of MSC by IFN-γ leading to the upregulation of CCL2 expression was essential for the suppressive effects, as administration of wild-type MSC into IFN-γ-deficient recipients, or IFN-γ receptor-deficient or CCL2-deficient MSC into wild-type mice failed to suppress airway inflammation. These results suggest that MSC activation by IFN-γ, followed by increased expression of CCL2 and recruitment of monocytes to the lungs, is essential for suppression by MSC in allergen-induced airway hyperresponsiveness and airway inflammation.
Collapse
Affiliation(s)
- Katsuyuki Takeda
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Tracy L Webb
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Fangkun Ning
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Yoshiki Shiraishi
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Daniel P Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Lyndah Chow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Mia J Smith
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Shigeru Ashino
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Amanda M Guth
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Sophie Hopkins
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Erwin W Gelfand
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, Denver, CO 80206; and
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|
40
|
Saradna A, Do DC, Kumar S, Fu QL, Gao P. Macrophage polarization and allergic asthma. Transl Res 2018; 191:1-14. [PMID: 29066321 PMCID: PMC5776696 DOI: 10.1016/j.trsl.2017.09.002] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Allergic asthma is associated with airway inflammation and airway hyperresponsiveness. Macrophage polarization has been shown to have a profound impact on asthma pathogenesis. On exposure to local microenvironments, recruited macrophages can be polarized into either classically activated (or M1) or alternatively activated (or M2) phenotypes. Macrophage polarization has been heavily associated with development of asthma. The process of regulation of macrophage polarization involves an intricate interplay between various cytokines, chemokines, transcriptional factors, and immune-regulatory cells. Different signals from the microenvironment are controlled by different receptors on the macrophages to initiate various macrophage polarization pathways. Most importantly, there is an increased attention on the epigenetic changes (eg, microRNAs, DNA methylation, and histone modification) that impact macrophage functional responses and M1/M2 polarization through modulating cellular signaling and signature gene expression. Thus, modulation of macrophage phenotypes through molecular intervention by targeting some of those potential macrophage regulators may have therapeutic potential in the treatment of allergic asthma and other allergic diseases. In this review, we will discuss the origin of macrophages, characterization of macrophages, macrophage polarization in asthma, and the underlying mechanisms regarding allergen-induced macrophage polarization with emphasis on the regulation of epigenetics, which will provide new insights into the therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Arjun Saradna
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Department of Internal Medicine, Maimonides Medical Center, Brooklyn, NY
| | - Danh C Do
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shruthi Kumar
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Bangalore Medical College and Research Institute, Bangalore, India
| | - Qing-Ling Fu
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peisong Gao
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
41
|
Li N, Hua J. Interactions between mesenchymal stem cells and the immune system. Cell Mol Life Sci 2017; 74:2345-2360. [PMID: 28214990 PMCID: PMC11107583 DOI: 10.1007/s00018-017-2473-5] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/24/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
In addition to being multi-potent, mesenchymal stem cells (MSCs) possess immunomodulatory functions that have been investigated as potential treatments in various immune disorders. MSCs can robustly interact with cells of the innate and adaptive immune systems, either through direct cell-cell contact or through their secretome. In this review, we discuss current findings regarding the interplay between MSCs and different immune cell subsets. We also draw attention to the mechanisms involved.
Collapse
Affiliation(s)
- Na Li
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
42
|
Moeinzadeh S, Shariati SRP, Kader S, Melero-Martin JM, Jabbari E. Devitalized Stem Cell Microsheets for Sustainable Release of Osteogenic and Vasculogenic Growth Factors and Regulation of Anti-Inflammatory Immune Response. ACTA ACUST UNITED AC 2017; 1. [PMID: 30221188 DOI: 10.1002/adbi.201600011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The objective of this work was to investigate the effect of devitalized human mesenchymal stem cells (hMSCs) and endothelial colony-forming cells (ECFCs) seeded on mineralized nanofiber microsheets on protein release, osteogenesis, vasculogenesis, and macrophage polarization. Calcium phosphate nanocrystals were grown on the surface of aligned, functionalized nanofiber microsheets. The microsheets were seeded with hMSCs, ECFCs, or a mixture of hMSCs+ECFCs, cultured for cell attachment, differentiated to the osteogenic or vasculogenic lineage, and devitalized by lyophilization. The release kinetic of total protein, bone morphogenetic protein-2 (BMP2), and vascular endothelial growth factor (VEGF) from the devitalized microsheets was measured. Next, hMSCs and/or ECFCs were seeded on the devitalized cell microsheets and cultured in the absence of osteo-/vasculo-inductive factors to determine the effect of devitalized cell microsheets on hMSC/ECFC differentiation. Human macrophages were seeded on the microsheets to determine the effect of devitalized cells on macrophage polarization. Based on the results, devitalized undifferentiated hMSC and vasculogenic-differentiated ECFC microsheets had highest sustained release of BMP2 and VEGF, respectively. The devitalized hMSC microsheets did not affect M2 macrophage polarization while vascular-differentiated, devitalized ECFC microsheets did not affect M1 polarization. Both groups stimulated higher M2 macrophage polarization compared to M1.
Collapse
Affiliation(s)
- Seyedsina Moeinzadeh
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Seyed Ramin Pajoum Shariati
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Safaa Kader
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA.,Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Esmaiel Jabbari
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
43
|
Hongyan L. Esculetin Attenuates Th2 and Th17 Responses in an Ovalbumin-Induced Asthmatic Mouse Model. Inflammation 2017; 39:735-43. [PMID: 26797918 DOI: 10.1007/s10753-015-0300-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The purpose of the current study was to investigate the anti-asthmatic effect of esculetin (ES) and explore its potential mechanism with a mouse model of allergic asthma. A total number of 50 mice were randomly assigned to five groups: control, model, dexamethasone (Dex, 2 mg/kg), and ES (20 mg/kg, 40 mg/kg). Mouse asthma model was developed with the sensitization and challenge of ovalbumin (OVA). The levels of IgE in serum, eosinophilia infiltration, Th2/Th17 cytokines, Th17 cell frequency, histological condition, and the protein expressions of RORγt, GATA3 were detected. Our study demonstrated that ES inhibited, OVA-induced eosinophil count, interleukin-4 (IL-4), IL-5, IL-13, and IL-17A levels were recovered in bronchoalveolar lavage fluid. Flow cytometry (FCM) studies revealed that ES substantially inhibited Th17 cells' percentage. Western blot study also indicated that ES downregulated RORγt and GATA3 expressions. Meanwhile, ES had beneficial effects on the histological alteration. These findings suggested that ES might effectively ameliorate the progression of asthma and could be used as a therapy for patients with allergic asthma.
Collapse
Affiliation(s)
- Long Hongyan
- Central Laboratory, Nanjing Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Nanjing University of T.C.M., 1 Jinling Road, Nanjing, 210001, Jiangsu Province, China.
| |
Collapse
|
44
|
Ellison B. Stem Cell Research and Social Justice: Aligning Scientific Progress with Social Need. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0063-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Sinclair KA, Yerkovich ST, Hopkins PMA, Chambers DC. Characterization of intercellular communication and mitochondrial donation by mesenchymal stromal cells derived from the human lung. Stem Cell Res Ther 2016; 7:91. [PMID: 27406134 PMCID: PMC4942965 DOI: 10.1186/s13287-016-0354-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/14/2016] [Accepted: 06/23/2016] [Indexed: 12/22/2022] Open
Abstract
Background Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are capable of repairing wounded lung epithelial cells by donating cytoplasmic material and mitochondria. Recently, we characterized two populations of human lung-derived mesenchymal stromal cells isolated from digested parenchymal lung tissue (LT-MSCs) from healthy individuals or from lung transplant recipients’ bronchoalveolar lavage fluid (BAL-MSCs). The aim of this study was to determine whether LT-MSCs and BAL-MSCs are also capable of donating cytoplasmic content and mitochondria to lung epithelial cells. Methods Cytoplasmic and mitochondrial transfer was assessed by co-culturing BEAS2B epithelial cells with Calcein AM or Mitotracker Green FM-labelled MSCs. Transfer was then measured by flow cytometry and validated by fluorescent microscopy. Molecular inhibitors were used to determine the contribution of microtubules/tunnelling nanotubes (TNTs, cytochalasin D), gap junctions (carbenoxolone), connexin-43 (gap26) and microvesicles (dynasore). Results F-actin microtubules/TNTs extending from BM-MSCs, LT-MSCs and BAL-MSCs to bronchial epithelial cells formed within 45 minutes of co-culturing cells. Each MSC population transferred a similar volume of cytoplasmic content to epithelial cells. Inhibiting microtubule/TNTs, gap junction formation and microvesicle endocytosis abrogated the transfer of cytoplasmic material from BM-MSCs, LT-MSCs and BAL-MSCs to epithelial cells. In contrast, blocking connexin-43 gap junction formation had no effect on cytoplasmic transfer. All MSC populations donated mitochondria to bronchial epithelial cells with similar efficiency. Mitochondrial transfer was reduced in all co-cultures after microtubule/TNT or endocytosis inhibition. Gap junction formation inhibition reduced mitochondrial transfer in BM-MSC and BAL-MSC co-cultures but had no effect on transfer in LT-MSC co-cultures. Connexin-43 inhibition did not impact mitochondrial transfer. Finally, bronchial epithelial cells were incapable of donating cytoplasmic content or mitochondria to any MSC population. Conclusion Similar to their bone marrow counterparts, LT-MSCs and BAL-MSCs can donate cytoplasmic content and mitochondria to bronchial epithelial cells via multiple mechanisms. Given that BM-MSCs utilize these mechanisms to mediate the repair of damaged bronchial epithelial cells, both LT-MSCs and BAL-MSCs will probably function similarly. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0354-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kenneth Andrew Sinclair
- School of Medicine, University of Queensland, Brisbane, QLD, Australia. .,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD, 4032, Australia.
| | - Stephanie Terase Yerkovich
- School of Medicine, University of Queensland, Brisbane, QLD, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD, 4032, Australia
| | - Peter Mark-Anthony Hopkins
- School of Medicine, University of Queensland, Brisbane, QLD, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD, 4032, Australia
| | - Daniel Charles Chambers
- School of Medicine, University of Queensland, Brisbane, QLD, Australia.,Queensland Lung Transplant Service, Ground Floor, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Chermside, Brisbane, QLD, 4032, Australia
| |
Collapse
|
46
|
Abstract
Lung macrophages link innate and adaptive immune responses during allergic airway inflammatory responses. Alveolar macrophages (AMs) and interstitial macrophages are two different phenotypes that differentially exert immunological function under physiological and pathological conditions. Exposure to pathogen induces polarization of AM cells into classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells). M1 cells dominantly express proinflammatory cytokines such as TNF-α and IL-1 β and induce lung inflammation and tissue damage. M2 cells are further divided into M2a and M2c subsets. M2a cells dominantly produce allergic cytokines IL-4 and IL-13, but M2c cells dominantly produce anti-inflammatory cytokine IL-10. M2a and M2c cells are differently involved in initiation, inflammation resolution, and tissue remodeling in the different stages of asthma. Microenvironment dynamically influences polarization of AM cells. Cytokines, chemokines, and immune-regulatory cells interplay and affect the balance between the polarization of M1 and M2 cells, subsequently influencing disease progression. Thus, modulation of AM phenotypes through molecular intervention has therapeutic potential in the treatment of asthma and other allergic inflammatory diseases. This review updated recent advances in polarization and functional specialization of these macrophage subtypes with emphasis on modulation of polarization of M2 cells in asthma of human subjects and animal models.
Collapse
Affiliation(s)
- Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
47
|
Mohammadian M, Sadeghipour HR, Kashani IR, Jahromi GP, Omidi A, Nejad AK, Golchoobian R, Boskabady MH. Evaluation of Simvastatin and Bone Marrow-Derived Mesenchymal Stem Cell Combination Therapy on Airway Remodeling in a Mouse Asthma Model. Lung 2016; 194:777-85. [PMID: 27161569 DOI: 10.1007/s00408-016-9884-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/22/2016] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The effect of bone marrow-derived mesenchymal stem cells (BMSCs) on asthma treatment was shown in our previous study. Several studies have shown the effect of statins on BMSC preservation and migration to sites of inflammation. In this study, the effects of simvastatin and BMSC combination therapy in an ovalbumin-induced asthma model in mouse were examined. METHODS Four groups of BALB/c mice were studied including control group (animals were not sensitized), asthma group (animals were sensitized by ovalbumin), asthma + simvastatin group (asthmatic animals were treated with simvastatin), and asthma + BMSC + simvastatin group (asthmatic animals were treated with simvastatin and BMSCs). BMSCs were isolated, characterized, labeled with BrdU, and transferred into asthmatic mice. BMSC migration, airways histopathology, and total and differential white blood cell (WBC) count in bronchoalveolar lavage (BAL) fluid were evaluated. RESULTS A significant increase in the number of BrdU-BMSCs was found in the lungs of mice treated with simvastatin + BMSCs compared to mice treated with BMSCs. The histopathological changes, BAL total WBC counts, and the percentage of neutrophils and eosinophils were increased in asthma group compared to the control group. Treatment with simvastatin significantly decreased airway inflammation and inflammatory cell infiltration. Combination therapy improved all measured parameters higher than simvastatin. Goblet cell hyperplasia and subepithelial fibrosis were also decreased in combination therapy group. CONCLUSION These results indicated that simvastatin and BMSC combination therapy was superior to simvastatin therapy and BMSC therapy alone in reduction of airway remodeling and lung inflammation in the ovalbumin-induced asthma model in mouse.
Collapse
Affiliation(s)
- Maryam Mohammadian
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Sadeghipour
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gila Pirzad Jahromi
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amene Omidi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kavian Nejad
- Department of Emergency Medical Services, Faculty of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ravie Golchoobian
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Boskabady
- Neurogenic Inflammation Research Center and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
48
|
Braza F, Dirou S, Forest V, Sauzeau V, Hassoun D, Chesné J, Cheminant-Muller MA, Sagan C, Magnan A, Lemarchand P. Mesenchymal Stem Cells Induce Suppressive Macrophages Through Phagocytosis in a Mouse Model of Asthma. Stem Cells 2016; 34:1836-45. [PMID: 26891455 DOI: 10.1002/stem.2344] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/19/2016] [Accepted: 02/01/2016] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell (MSC) immunosuppressive functions make them attractive candidates for anti-inflammatory therapy in allergic asthma. However, the mechanisms by which they ensure therapeutic effects remain to be elucidated. In an acute mouse model of house dust mite (Der f)-induced asthma, one i.v. MSC injection was sufficient to normalize and stabilize lung function in Der f-sensitized mice as compared to control mice. MSC injection decreased in vivo airway responsiveness and decreased ex vivo carbachol-induced bronchial contraction, maintaining bronchial expression of the inhibitory type 2 muscarinic receptor. To evaluate in vivo MSC survival, MSCs were labeled with PKH26 fluorescent marker prior to i.v. injection, and 1 to 10 days later total lungs were digested to obtain single-cell suspensions. 91.5 ± 2.3% and 86.6 ± 6.3% of the recovered PKH26(+) lung cells expressed specific macrophage markers in control and Der f mice, respectively, suggesting that macrophages had phagocyted in vivo the injected MSCs. Interestingly, only PKH26(+) macrophages expressed M2 phenotype, while the innate PKH26(-) macrophages expressed M1 phenotype. Finally, the remaining 0.5% PKH26(+) MSCs expressed 10- to 100-fold more COX-2 than before injection, suggesting in vivo MSC phenotype modification. Together, the results of this study indicate that MSCs attenuate asthma by being phagocyted by lung macrophages, which in turn acquire a M2 suppressive phenotype. Stem Cells 2016;34:1836-1845.
Collapse
Affiliation(s)
- Faouzi Braza
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Stéphanie Dirou
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France.,CHU de Nantes, Nantes, F-44000, France
| | - Virginie Forest
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Vincent Sauzeau
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Dorian Hassoun
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Julie Chesné
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Marie-Aude Cheminant-Muller
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France
| | - Christine Sagan
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France.,CHU de Nantes, Nantes, F-44000, France
| | - Antoine Magnan
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France.,CHU de Nantes, Nantes, F-44000, France
| | - Patricia Lemarchand
- INSERM, UMR1087, l'institut du thorax, Nantes, F-44000, France.,CNRS, UMR 6291, Nantes, F-44000, France.,Université de Nantes, Nantes, F-44000, France.,CHU de Nantes, Nantes, F-44000, France
| |
Collapse
|
49
|
Hsu YC, Wu YT, Yu TH, Wei YH. Mitochondria in mesenchymal stem cell biology and cell therapy: From cellular differentiation to mitochondrial transfer. Semin Cell Dev Biol 2016; 52:119-31. [PMID: 26868759 DOI: 10.1016/j.semcdb.2016.02.011] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are characterized to have the capacity of self-renewal and the potential to differentiate into mesoderm, ectoderm-like and endoderm-like cells. MSCs hold great promise for cell therapies due to their multipotency in vitro and therapeutic advantage of hypo-immunogenicity and lower tumorigenicity. Moreover, it has been shown that MSCs can serve as a vehicle to transfer mitochondria into cells after cell transplantation. Mitochondria produce most of the energy through oxidative phosphorylation in differentiated cells. It has been increasingly clear that the switch of energy supply from glycolysis to aerobic metabolism is essential for successful differentiation of MSCs. Post-translational modifications of proteins have been established to regulate mitochondrial function and metabolic shift during MSCs differentiation. In this article, we review and provide an integrated view on the roles of different protein kinases and sirtuins in the maintenance and differentiation of MSCs. Importantly, we provide evidence to suggest that alteration in the expression of Sirt3 and Sirt5 and relative changes in the acylation levels of mitochondrial proteins might be involved in the activation of mitochondrial function and adipogenic differentiation of adipose-derived MSCs. We summarize their roles in the regulation of mitochondrial biogenesis and metabolism, oxidative responses and differentiation of MSCs. On the other hand, we discuss recent advances in the study of mitochondrial dynamics and mitochondrial transfer as well as their roles in the differentiation and therapeutic application of MSCs to improve cell function in vitro and in animal models. Accumulating evidence has substantiated that the therapeutic potential of MSCs is conferred not only by cell replacement and paracrine effects but also by transferring mitochondria into injured tissues or cells to modulate the cellular metabolism in situ. Therefore, elucidation of the underlying mechanisms in the regulation of mitochondrial metabolism of MSCs may ultimately improve therapeutic outcomes of stem cell therapy in the future.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan
| | - Yu-Ting Wu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ting-Hsien Yu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Yau-Huei Wei
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
50
|
Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, Kao WWY. Extrinsic and Intrinsic Mechanisms by Which Mesenchymal Stem Cells Suppress the Immune System. Ocul Surf 2016; 14:121-34. [PMID: 26804815 DOI: 10.1016/j.jtos.2015.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/12/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are a group of fibroblast-like multipotent mesenchymal stromal cells that have the ability to differentiate into osteoblasts, adipocytes, and chondrocytes. Recent studies have demonstrated that MSCs possess a unique ability to exert suppressive and regulatory effects on both adaptive and innate immunity in an autologous and allogeneic manner. A vital step in stem cell transplantation is overcoming the potential graft-versus-host disease, which is a limiting factor to transplantation success. Given that MSCs attain powerful differentiation capabilities and also present immunosuppressive properties, which enable them to survive host immune rejection, MSCs are of great interest. Due to their ability to differentiate into different cell types and to suppress and modulate the immune system, MSCs are being developed for treating a plethora of diseases, including immune disorders. Moreover, in recent years, MSCs have been genetically engineered to treat and sometimes even cure some diseases, and the use of MSCs for cell therapy presents new perspectives for overcoming tissue rejection. In this review, we discuss the potential extrinsic and intrinsic mechanisms that underlie MSCs' unique ability to modulate inflammation, and both innate and adaptive immunity.
Collapse
Affiliation(s)
- Vivien J Coulson-Thomas
- Department of Ophthalmology, University of Cincinnati, Ohio, USA; John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
| | | | | | - Winston W-Y Kao
- Department of Ophthalmology, University of Cincinnati, Ohio, USA.
| |
Collapse
|