Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells

Cells - Tập 9 Số 7 - Trang 1660
Claudia Terraza‐Aguirre1,2, Mauricio Campos‐Mora1,3, Roberto Elizondo‐Vega4, Rafaël Contreras1,2, Patricia Luz‐Crawford5, Christian Jørgensen1,2, Farida Djouad1,2
1CHRU Montpellier - Centre Hospitalier Régional Universitaire [Montpellier] (191 avenue du doyen Gaston Giraud 34295 Montpellier - France)
2Cellules Souches, Plasticité Cellulaire, Médecine Régénératrice et Immunothérapies (IRMB) (Institute for Regenerative Medicine and Biotherapy - 80 rue Augustin Fliche 34295 Montpellier - Cedex 5 - France)
3IGH - Institut de génétique humaine (institut de Génétique humaine 141 Rue de la Cardonille 34396 MONTPELLIER CEDEX 5 - France)
4Universidad de Concepción - University of Concepcion [Chile] (Concepción, Bio Bio, Chie - Chile)
5UANDES - Universidad de los Andes [Santiago] (Monseñor Álvaro del Portillo 12455, Santiago, Las Condes - Chili - Chile)

Tóm tắt

Mesenchymal stem cells (MSCs) exhibit potent immunoregulatory abilities by interacting with cells of the adaptive and innate immune system. In vitro, MSCs inhibit the differentiation of T cells into T helper 17 (Th17) cells and repress their proliferation. In vivo, the administration of MSCs to treat various experimental inflammatory and autoimmune diseases, such as rheumatoid arthritis, type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, and bowel disease showed promising therapeutic results. These therapeutic properties mediated by MSCs are associated with an attenuated immune response characterized by a reduced frequency of Th17 cells and the generation of regulatory T cells. In this manuscript, we review how MSC and Th17 cells interact, communicate, and exchange information through different ways such as cell-to-cell contact, secretion of soluble factors, and organelle transfer. Moreover, we discuss the consequences of this dynamic dialogue between MSC and Th17 well described by their phenotypic and functional plasticity.

Từ khóa


Tài liệu tham khảo

Friedenstein, 1968, Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues, Transplantation, 6, 230, 10.1097/00007890-196803000-00009

Owen, 1988, Stromal stem cells: Marrow-derived osteogenic precursors, Ciba Found. Symp., 136, 42

Friedenstein, 1987, Bone marrow osteogenic stem cells: In vitro cultivation and transplantation in diffusion chambers, Cell Tissue Kinet., 20, 263

Piersma, 1985, Characterization of fibroblastic stromal cells from murine bone marrow, Exp. Hematol., 13, 237

Caplan, 1991, Mesenchymal stem cells, J. Orthop. Res., 9, 641, 10.1002/jor.1100090504

Romanov, 2005, Mesenchymal stem cells from human bone marrow and adipose tissue: Isolation, characterization, and differentiation potentialities, Bull. Exp. Biol. Med., 140, 138, 10.1007/s10517-005-0430-z

Kassis, 2006, Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads, Bone Marrow Transplant., 37, 967, 10.1038/sj.bmt.1705358

Cuenca, 2015, Characterization of menstrual stem cells: Angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells, Stem Cell Res. Ther., 6, 32, 10.1186/s13287-015-0013-5

Djouad, 2005, Transcriptional Profiles Discriminate Bone Marrow-Derived and Synovium-Derived Mesenchymal Stem Cells, Arthritis Res. Ther., 7, R1304, 10.1186/ar1827

Tylzanowski, 2001, Multipotent Mesenchymal Stem Cells from Adult Human Synovial Membrane, Arthritis Rheum., 44, 1928, 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P

Huang, 2010, Stem/Progenitor Cell-Mediated De Novo Regeneration of Dental Pulp with Newly Deposited Continuous Layer of Dentin in an In Vivo Model, Tissue Eng. Part A, 16, 605, 10.1089/ten.tea.2009.0518

Erices, 2003, Human Cord Blood-Derived Mesenchymal Stem Cells Home and Survive in the Marrow of Immunodeficient Mice after Systemic Infusion, Cell Transplant., 12, 555, 10.3727/000000003108747154

Miao, 2006, Isolation of mesenchymal stem cells from human placenta: Comparison with human bone marrow mesenchymal stem cells, Cell Biol. Int., 30, 681, 10.1016/j.cellbi.2006.03.009

Weiss, 2008, Immune Properties of Human Umbilical Cord Wharton’s Jelly-Derived Cells, Stem Cells, 26, 2865, 10.1634/stemcells.2007-1028

Marongiu, 2010, Isolation of amniotic mesenchymal stem cells, Curr. Protoc. Stem Cell Biol., 12, 1E.5.1, 10.1002/9780470151808.sc01e05s12

Dominici, 2006, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement, Cytotherapy, 8, 315, 10.1080/14653240600855905

Mhaske, 2017, Interleukin-3 enhances the migration of human mesenchymal stem cells by regulating expression of CXCR4, Stem Cell Res. Ther., 8, 168, 10.1186/s13287-017-0618-y

Nishikawa, 2019, Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression via CCR5, Cell Death Dis., 10, 264, 10.1038/s41419-019-1508-2

Lamoury, 2007, Human mesenchymal stem cells constitutively express chemokines and chemokine receptors that can be upregulated by cytokines, IFN-beta, and Copaxone, J. Interferon Cytokine Res., 27, 53, 10.1089/jir.2006.0037

Sordi, 2005, Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets, Blood, 106, 419, 10.1182/blood-2004-09-3507

Li, 2015, Bone Marrow Mesenchymal Stem Cells Protect Alveolar Macrophages from Lipopolysaccharide-Induced Apoptosis Partially by Inhibiting the Wnt/β-Catenin Pathway, Cell Biol. Int., 39, 192, 10.1002/cbin.10359

Qu, 2007, Anti-apoptotic Effects of Mesenchymal Stem Cells on Cardiac Myocytes: In Vitro Study with Rats, Zhonghua Yi Xue Za Zhi, 87, 271

Huang, 2015, Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated with GFAP Downregulation after Ischemic Stroke via p38 MAPK and JNK, J. Cell. Physiol., 230, 2461, 10.1002/jcp.24981

Tang, 2005, Paracrine Action Enhances the Effects of Autologous Mesenchymal Stem Cell Transplantation on Vascular Regeneration in Rat Model of Myocardial Infarction, Ann. Thorac. Surg., 80, 229, 10.1016/j.athoracsur.2005.02.072

Ucuzian, 2010, Molecular Mediators of Angiogenesis, J. Burn Care Res., 31, 158, 10.1097/BCR.0b013e3181c7ed82

Kang, 2008, Engineering robust and functional vascular networks in vivo with human adult and cord blood-derived progenitor cells, Circ. Res., 103, 194, 10.1161/CIRCRESAHA.108.178590

Lin, 2012, Equal modulation of endothelial cell function by four distinct tissue-specific mesenchymal stem cells, Angiogenesis, 15, 443, 10.1007/s10456-012-9272-2

Traktuev, 2009, Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells, Circ. Res., 104, 1410, 10.1161/CIRCRESAHA.108.190926

Kachgal, 2011, Mesenchymal stem cells from adipose and bone marrow promote angiogenesis via distinct cytokine and protease expression mechanisms, Angiogenesis, 14, 47, 10.1007/s10456-010-9194-9

Roubelakis, M.G., Tsaknakis, G., Pappa, K.I., Anagnou, N.P., and Watt, S.M. (2013). Spindle Shaped Human Mesenchymal Stem/Stromal Cells from Amniotic Fluid Promote Neovascularization. PLoS ONE, 8.

Gollahalli, 2010, Adipose tissue progenitor cells directly interact with endothelial cells to induce vascular network formation, Tissue Eng. Part A, 16, 2953, 10.1089/ten.tea.2009.0635

Kwon, 2014, Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis, Vasc. Pharm., 63, 19, 10.1016/j.vph.2014.06.004

Razban, 2012, HIF-1α Overexpression Induces Angiogenesis in Mesenchymal Stem Cells, BioRes. Open Access, 1, 174, 10.1089/biores.2012.9905

Ranganath, 2012, Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease, Cell Stem Cell, 10, 244, 10.1016/j.stem.2012.02.005

Liu, 2013, Hypoxia preconditioned human adipose derived mesenchymal stem cells enhance angiogenic potential via secretion of increased VEGF and bFGF, Cell Biol. Int., 37, 551, 10.1002/cbin.10097

Kramann, 2017, Mesenchymal Stem Cells in Fibrotic Disease, Cell Stem Cell, 21, 166, 10.1016/j.stem.2017.07.011

Rockel, 2020, Anti-Fibrotic Mechanisms of Exogenously-Expanded Mesenchymal Stromal Cells for Fibrotic Diseases, Semin. Cell Dev. Biol., 101, 87, 10.1016/j.semcdb.2019.10.014

Firestein, 2003, Evolving Concepts of Rheumatoid Arthritis, Nature, 423, 356, 10.1038/nature01661

Ospelt, 2007, Cells of the Synovium in Rheumatoid Arthritis. Synovial Fibroblasts, Arthritis Res. Ther., 9, 223, 10.1186/ar2337

Yang, Z., Shen, Y., Oishi, H., Matteson, E.L., Tian, L., Goronzy, J.J., and Weyand, C.M. (2016). Restoring Oxidant Signaling Suppresses Proarthritogenic T Cell Effector Functions in Rheumatoid Arthritis. Sci. Transl. Med., 8.

Torres, 2019, Mesenchymal Stem Cells Improve Rheumatoid Arthritis Progression by Controlling Memory T Cell Response, Front. Immunol., 10, 798, 10.3389/fimmu.2019.00798

Ehrenstein, 2004, Compromised Function of Regulatory T Cells in Rheumatoid Arthritis and Reversal by Anti-TNFalpha Therapy, J. Exp. Med., 200, 277, 10.1084/jem.20040165

Richardson, 2013, Novel Immunotherapies for Rheumatoid Arthritis, Clin. Med., 13, 391, 10.7861/clinmedicine.13-4-391

Luz-Crawford, P., Noël, D., Fernandez, X., Khoury, M., Figueroa, F., Carrión, F., Jorgensen, C., and Djouad, F. (2012). Mesenchymal Stem Cells Repress Th17 Molecular Program through the PD-1 Pathway. PLoS ONE, 7.

Tejedor, 2015, Glucocorticoid-induced leucine zipper governs the therapeutic potential of mesenchymal stem cells by inducing a switch from pathogenic to regulatory Th17 cells in a mouse model of collagen-induced arthritis, Arthritis Rheumatol., 67, 1514, 10.1002/art.39069

Jorgensen, 2017, Mesenchymal Stem Cells Direct the Immunological Fate of Macrophages, Macrophages, 62, 61, 10.1007/978-3-319-54090-0_4

Hernandez, 2019, Mesenchymal Stem Cell Repression of Th17 Cells Is Triggered by Mitochondrial Transfer, Stem Cell Res. Ther., 10, 232, 10.1186/s13287-019-1307-9

Rozenberg, 2016, Human Mesenchymal Stem Cells Impact Th17 and Th1 Responses Through a Prostaglandin E2 and Myeloid-Dependent Mechanism, Stem Cells Transl. Med., 5, 1506, 10.5966/sctm.2015-0243

Leyendecker, 2018, The Use of Human Mesenchymal Stem Cells as Therapeutic Agents for the In Vivo Treatment of Immune-Related Diseases: A Systematic Review, Front. Immunol., 9, 2056, 10.3389/fimmu.2018.02056

Li, 2020, Mesenchymal Stem Cells Induced Regulatory Dendritic Cells from Hemopoietic Progenitor Cells through Notch Pathway and TGF-β Synergistically, Immunol. Lett., 222, 49, 10.1016/j.imlet.2020.03.005

Chen, 2015, Effects of Mesenchymal Stromal Cells on Human Myeloid Dendritic Cell Differentiation and Maturation in a Humanized Mouse Model, J. Immunol. Methods, 427, 100, 10.1016/j.jim.2015.10.008

Chen, 2019, Mesenchymal Stem Cells Alleviate Moderate-to-Severe Psoriasis by Reducing the Production of Type I Interferon (IFN-I) by Plasmacytoid Dendritic Cells (pDCs), Stem Cells Int., 2019, 6961052, 10.1155/2019/6961052

Lu, 2019, Mesenchymal Stem Cells Induce Dendritic Cell Immune Tolerance via Paracrine Hepatocyte Growth Factor to Alleviate Acute Lung Injury, Stem Cell Res. Ther., 10, 372, 10.1186/s13287-019-1488-2

Favaro, 2016, Human Mesenchymal Stem Cells and Derived Extracellular Vesicles Induce Regulatory Dendritic Cells in Type 1 Diabetic Patients, Diabetologia, 59, 325, 10.1007/s00125-015-3808-0

He, 2019, MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing, Stem Cells Int., 2019, 7132708, 10.1155/2019/7132708

Liu, 2019, MSC-secreted TGF-β Regulates Lipopolysaccharide-Stimulated Macrophage M2-like Polarization via the Akt/FoxO1 Pathway, Stem Cell Res. Ther., 10, 34, 10.1186/s13287-019-1447-y

Morrison, 2017, Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer, Am. J. Respir. Crit. Care Med., 196, 1275, 10.1164/rccm.201701-0170OC

Akiyama, 2012, Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis, Cell Stem Cell, 10, 544, 10.1016/j.stem.2012.03.007

Murphy, 2019, TNF-α/IL-1β-licensed Mesenchymal Stromal Cells Promote Corneal Allograft Survival via Myeloid Cell-Mediated Induction of Foxp3 + Regulatory T Cells in the Lung, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol., 33, 9404

Braga, 2017, Mesenchymal Stromal Cell-Derived Microvesicles Regulate an Internal Pro-Inflammatory Program in Activated Macrophages, Front. Immunol., 8, 881, 10.3389/fimmu.2017.00881

Zhu, 2014, Human Mesenchymal Stem Cell Microvesicles for Treatment of Escherichia Coli Endotoxin-Induced Acute Lung Injury in Mice, Stem Cells, 32, 116, 10.1002/stem.1504

Jackson, 2016, Mitochondrial Transfer via Tunneling Nanotubes Is an Important Mechanism by Which Mesenchymal Stem Cells Enhance Macrophage Phagocytosis in the In Vitro and In Vivo Models of ARDS, Stem Cells, 34, 2210, 10.1002/stem.2372

Hu, 2019, Differential Immunomodulatory Effects of Human Bone Marrow-Derived Mesenchymal Stromal Cells on Natural Killer Cells, Stem Cells Dev., 28, 933, 10.1089/scd.2019.0059

Lu, 2015, TLR4 Plays a Crucial Role in MSC-induced Inhibition of NK Cell Function, Biochem. Biophys. Res. Commun., 464, 541, 10.1016/j.bbrc.2015.07.002

Blanco, 2016, Immunomodulatory Effects of Bone Marrow versus Adipose Tissue-Derived Mesenchymal Stromal Cells on NK Cells: Implications in the Transplantation Setting, Eur. J. Haematol., 97, 528, 10.1111/ejh.12765

Bassi, 2016, Differential and Transferable Modulatory Effects of Mesenchymal Stromal Cell-Derived Extracellular Vesicles on T, B and NK Cell Functions, Sci. Rep., 6, 24120, 10.1038/srep24120

Alves, V.B.F., de Sousa, B.C., Fonseca, M.T.C., Ogata, H., Caliári-Oliveira, C., Yaochite, J.N.U., Rodrigues Júnior, V., Chica, J.E.L., da Silva, J.S., and Malmegrim, K.C.R. (2019). A Single Administration of Human Adipose Tissue-Derived Mesenchymal Stromal Cells (MSC) Induces Durable and Sustained Long-Term Regulation of Inflammatory Response in Experimental Colitis. Clin. Exp. Immunol., 196.

Munneke, 2018, Mesenchymal Stromal Cells Stimulate the Proliferation and IL-22 Production of Group 3 Innate Lymphoid Cells, J. Immunol., 201, 1165, 10.4049/jimmunol.1700901

Lu, 2019, B Lymphocytes Are the Target of Mesenchymal Stem Cells Immunoregulatory Effect in a Murine Graft-versus-Host Disease Model, Cell Transplant., 28, 1279, 10.1177/0963689719860127

Luk, 2017, Inflammatory Conditions Dictate the Effect of Mesenchymal Stem or Stromal Cells on B Cell Function, Front. Immunol., 8, 1042, 10.3389/fimmu.2017.01042

Wang, 2014, Requirement of B7-H1 in Mesenchymal Stem Cells for Immune Tolerance to Cardiac Allografts in Combination Therapy with Rapamycin, Transpl. Immunol., 31, 65, 10.1016/j.trim.2014.06.005

Cho, 2017, Mesenchymal stem cells ameliorate B-cell-mediated immune responses and increase IL-10-expressing regulatory B cells in an EBI3-dependent manner, Cell. Mol. Immunol., 14, 895, 10.1038/cmi.2016.59

Gupte, 2017, In-Vitro Generation of Interleukin-10 Secreting B-Regulatory Cells From Donor Adipose Tissue Derived Mesenchymal Stem Cells and Recipient Peripheral Blood Mononuclear Cells for Potential Cell Therapy, Biomed. J., 40, 49, 10.1016/j.bj.2017.01.003

Franquesa, 2015, Human Adipose Tissue-Derived Mesenchymal Stem Cells Abrogate Plasmablast Formation and Induce Regulatory B Cells Independently of T Helper Cells, Stem Cells, 33, 880, 10.1002/stem.1881

Jang, 2020, Metformin Enhances the Immunomodulatory Potential of Adipose-Derived Mesenchymal Stem Cells through STAT1 in an Animal Model of Lupus, Rheumatology, 59, 1426, 10.1093/rheumatology/kez631

Kiernan, 2020, Allogeneic Chondrogenic Mesenchymal Stromal Cells alter Helper T Cell Subsets in CD4+ Memory T Cells, Tissue Eng. Part A, 26, 490, 10.1089/ten.tea.2019.0177

Lee, K., Park, N., Jung, H., Rim, Y.A., Nam, Y., Lee, J., Park, S.H., and Ju, J.H. (2018). Mesenchymal Stem Cells Ameliorate Experimental Arthritis via Expression of interleukin-1 Receptor Antagonist. PLoS ONE, 13.

Pianta, 2015, Amniotic Membrane Mesenchymal Cells-Derived Factors Skew T Cell Polarization Toward Treg and Downregulate Th1 and Th17 Cells Subsets, Stem Cell Rev. Rep., 11, 394, 10.1007/s12015-014-9558-4

Zhao, 2019, Human Gingiva Tissue-Derived MSC Ameliorates Immune-Mediated Bone Marrow Failure of Aplastic Anemia via Suppression of Th1 and Th17 Cells and Enhancement of CD4+Foxp3+ Regulatory T Cells Differentiation, Am. J. Transl. Res., 11, 7627

Ng, 2016, Immunomodulatory Properties of Induced Pluripotent Stem Cell-Derived Mesenchymal Cells, J. Cell. Biochem., 117, 2844, 10.1002/jcb.25596

Cosenza, 2018, Mesenchymal Stem Cells-Derived Exosomes Are More Immunosuppressive Than Microparticles in Inflammatory Arthritis, Theranostics, 8, 1399, 10.7150/thno.21072

Luciano, 2015, Immunoregulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles on T Lymphocytes, Cell Transplant., 24, 2615, 10.3727/096368915X687543

Laso-García, F., Ramos-Cejudo, J., Carrillo-Salinas, F., Otero-Ortega, L., Feliú, A., Gómez-de Frutos, M., Mecha, M., Díez-Tejedor, E., Guaza, C., and Gutiérrez-Fernández, M. (2018). Therapeutic Potential of Extracellular Vesicles Derived from Human Mesenchymal Stem Cells in a Model of Progressive Multiple Sclerosis. PLoS ONE, 13.

Oh, 2017, MSC-Derived Extracellular Vesicles Attenuate Immune Responses in Two Autoimmune Murine Models: Type 1 Diabetes and Uveoretinitis, Stem Cell Rep., 8, 1214, 10.1016/j.stemcr.2017.04.008

Court, 2020, Mitochondrial Transfer from MSCs to T Cells Induces Treg Differentiation and Restricts Inflammatory Response, EMBO Rep., 21, e48052, 10.15252/embr.201948052

Bai, 2009, Human Bone Marrow-Derived Mesenchymal Stem Cells Induce Th2-polarized Immune Response and Promote Endogenous Repair in Animal Models of Multiple Sclerosis, Glia, 57, 1192, 10.1002/glia.20841

Hur, 2020, Evaluation of Human MSCs Treatment Frequency on Airway Inflammation in a Mouse Model of Acute Asthma, J. Korean Med. Sci., 35, e188, 10.3346/jkms.2020.35.e188

Fang, 2020, Small Extracellular Vesicles Derived from Human MSCs Prevent Allergic Airway Inflammation via Immunomodulation on Pulmonary Macrophages, Cell Death Dis., 11, 409, 10.1038/s41419-020-2606-x

Chan, 2016, The Modulation of Th2 Immune Pathway in the Immunosuppressive Effect of Human Umbilical Cord Mesenchymal Stem Cells in a Murine Asthmatic Model, Inflamm. Res. Off. J. Eur. Histamine Res. Soc., 65, 795

Patel, 2014, Treg/Th17 Polarization by Distinct Subsets of Breast Cancer Cells Is Dictated by the Interaction with Mesenchymal Stem Cells, J. Cancer Stem Cell Res., 2014, e1003

Geng, 2020, Reduced Let-7f in Bone Marrow-Derived Mesenchymal Stem Cells Triggers Treg/Th17 Imbalance in Patients with Systemic Lupus Erythematosus, Front. Immunol., 11, 233, 10.3389/fimmu.2020.00233

Cao, 2020, Therapeutic Effect of Mesenchymal Stem Cell on Hashimoto’s Thyroiditis in a Rat Model by Modulating Th17/Treg Cell Balance, Autoimmunity, 53, 35, 10.1080/08916934.2019.1697689

Wang, 2017, Gastric Cancer Tissue-Derived Mesenchymal Stem Cells Impact Peripheral Blood Mononuclear Cells via Disruption of Treg/Th17 Balance to Promote Gastric Cancer Progression, Exp. Cell Res., 361, 19, 10.1016/j.yexcr.2017.09.036

Chen, 2016, Immunomodulatory Effects of Mesenchymal Stromal Cells-Derived Exosome, Immunol. Res., 64, 831, 10.1007/s12026-016-8798-6

Roura, 2017, Nanosized UCMSC-Derived Extracellular Vesicles but Not Conditioned Medium Exclusively Inhibit the Inflammatory Response of Stimulated T Cells: Implications for Nanomedicine, Theranostics, 7, 270, 10.7150/thno.16154

Wang, 2017, The Regulation of the Treg/Th17 Balance by Mesenchymal Stem Cells in Human Systemic Lupus Erythematosus, Cell. Mol. Immunol., 14, 423, 10.1038/cmi.2015.89

Yu, 2016, High expression of heme oxygenase-1 in target organs may attenuate acute graft-versus-host disease through regulation of immune balance of TH17/Treg, Transpl. Immunol., 37, 10, 10.1016/j.trim.2016.05.002

Yang, 2018, Hydrogen sulfide promotes immunomodulation of gingiva-derived mesenchymal stem cells via the Fas/FasL coupling pathway, Stem Cell Res. Ther., 9, 62, 10.1186/s13287-018-0804-6

Yu, 2019, Acetylsalicylic acid rescues the immunomodulation of inflamed gingiva-derived mesenchymal stem cells via upregulating FasL in mice, Stem Cell Res. Ther., 10, 368, 10.1186/s13287-019-1485-5

Azevedo, R.I., Minskaia, E., Fernandes-Platzgummer, A., Vieira, A.I.S., da Silva, C.L., Cabral, J.M.S., and Lacerda, J.F. (2020). Mesenchymal Stromal Cells Induce Regulatory T Cells via Epigenetic Conversion of Human Conventional CD4 T Cells In Vitro. Stem Cells.

Djouad, 2007, Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism, Stem Cells, 25, 2025, 10.1634/stemcells.2006-0548

Gao, 2017, Effects of Mesenchymal Stem Cells from Human Induced Pluripotent Stem Cells on Differentiation, Maturation, and Function of Dendritic Cells, Stem Cell Res. Ther., 8, 48, 10.1186/s13287-017-0499-0

Abumaree, 2013, Human Placental Mesenchymal Stem Cells (pMSCs) Play a Role as Immune Suppressive Cells by Shifting Macrophage Differentiation from Inflammatory M1 to Anti-Inflammatory M2 Macrophages, Stem Cell Rev. Rep., 9, 620, 10.1007/s12015-013-9455-2

Fan, L., Hu, C., Chen, J., Cen, P., Wang, J., and Li, L. (2016). Interaction between Mesenchymal Stem Cells and B-Cells. Int. J. Mol. Sci., 17.

Miossec, 2009, Interleukin-17 and Type 17 Helper T Cells, N. Engl. J. Med., 361, 888, 10.1056/NEJMra0707449

Yasuda, 2019, The pathogenicity of Th17 cells in autoimmune diseases, Semin. Immunopathol., 41, 283, 10.1007/s00281-019-00733-8

Lee, G.R. (2018). The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci., 19.

Ueno, 2018, Th17 Plasticity and Its Relevance to Inflammatory Bowel Disease, J. Autoimmun., 87, 38, 10.1016/j.jaut.2017.12.004

Ren, 2017, The Functional Stability of FOXP3 and RORγt in Treg and Th17 and Their Therapeutic Applications, Adv. Protein Chem. Struct. Biol., 107, 155, 10.1016/bs.apcsb.2016.10.002

Harris, 2002, Prostaglandins as modulators of immunity, Trends Immunol., 23, 144, 10.1016/S1471-4906(01)02154-8

Yao, 2009, Prostaglandin E2-EP4 signaling promotes immune inflammation through Th1 cell differentiation and Th17 cell expansion, Nat. Med., 15, 633, 10.1038/nm.1968

Boniface, 2009, Prostaglandin E2 regulates Th17 cell differentiation and function through cyclic AMP and EP2/EP4 receptor signaling, J. Exp. Med., 206, 535, 10.1084/jem.20082293

Duffy, 2011, Mesenchymal stem cell inhibition of T-helper 17 cell-differentiation is triggered by cell-cell contact and mediated by prostaglandin E2 via the EP4 receptor, Eur. J. Immunol., 41, 2840, 10.1002/eji.201141499

Kalinski, 2012, Regulation of immune responses by prostaglandin E2, J. Immunol., 188, 21, 10.4049/jimmunol.1101029

Vasilev, 2019, Secretory Factors Produced by Adipose Mesenchymal Stem Cells Downregulate Th17 and Increase Treg Cells in Peripheral Blood Mononuclear Cells from Rheumatoid Arthritis Patients, Rheumatol. Int., 39, 819, 10.1007/s00296-019-04296-7

Qu, 2012, Mesenchymal Stem Cells Inhibit Th17 Cell Differentiation by IL-10 Secretion, Exp. Hematol., 40, 761, 10.1016/j.exphem.2012.05.006

Najar, 2019, Th17 Immune Response to Adipose Tissue-Derived Mesenchymal Stromal Cells, J. Cell. Physiol., 234, 21145, 10.1002/jcp.28717

Svobodova, 2012, The Role of Mouse Mesenchymal Stem Cells in Differentiation of Naive T-cells into Anti-Inflammatory Regulatory T-cell or Proinflammatory Helper T-cell 17 Population, Stem Cells Dev., 21, 901, 10.1089/scd.2011.0157

Li, 2017, Human iPSC-MSC-Derived Xenografts Modulate Immune Responses by Inhibiting the Cleavage of Caspases, Stem Cells, 35, 1719, 10.1002/stem.2638

Bouffi, C., Bony, C., Courties, G., Jorgensen, C., and Noël, D. (2010). IL-6-Dependent PGE2 Secretion by Mesenchymal Stem Cells Inhibits Local Inflammation in Experimental Arthritis. PLoS ONE, 5.

Harrell, 2020, The Role of Interleukin 1 Receptor Antagonist in Mesenchymal Stem Cell-Based Tissue Repair and Regeneration, BioFactors, 46, 263, 10.1002/biof.1587

Ren, 2009, Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression, Stem Cells, 27, 1954, 10.1002/stem.118

Su, 2014, Phylogenetic distinction of iNOS and IDO function in mesenchymal stem cell-mediated immunosuppression in mammalian species, Cell Death Differ., 21, 388, 10.1038/cdd.2013.149

Volarevic, 2019, Galectin 3 Protects from Cisplatin-Induced Acute Kidney Injury by Promoting TLR-2-Dependent Activation of IDO1/Kynurenine Pathway in Renal DCs, Theranostics, 9, 5976, 10.7150/thno.33959

Yang, 2019, Indoleamine 2,3-Dioxygenase (IDO) Regulates Th17/Treg Immunity in Experimental IgA Nephropathy, Folia Biol., 65, 101, 10.14712/fb2019065020101

Routy, 2016, The Kynurenine Pathway Is a Double-Edged Sword in Immune-Privileged Sites and in Cancer: Implications for Immunotherapy, Int. J. Tryptophan Res., 9, 67, 10.4137/IJTR.S38355

Milosavljevic, 2018, Mesenchymal stem cells attenuate liver fibrosis by suppressing Th17 cells–An experimental study, Transpl. Int., 31, 102, 10.1111/tri.13023

Dulak, 2012, Haem oxygenase-1: Non-canonical roles in physiology and pathology, Clin. Sci., 122, 93, 10.1042/CS20110147

Shen, 2017, Immunomodulatory Effects of Bone Marrow Mesenchymal Stem Cells Overexpressing Heme Oxygenase-1: Protective Effects on Acute Rejection Following Reduced-Size Liver Transplantation in a Rat Model, Cell. Immunol., 313, 10, 10.1016/j.cellimm.2016.12.006

Langrzyk, 2018, Critical View on Mesenchymal Stromal Cells in Regenerative Medicine, Antioxid. Redox Signal., 29, 169, 10.1089/ars.2017.7159

Ren, 2008, Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide, Cell Stem Cell, 2, 141, 10.1016/j.stem.2007.11.014

Niedbala, 2006, Role of Nitric Oxide in the Regulation of T Cell Functions, Ann. Rheum. Dis., 65, iii37, 10.1136/ard.2006.058446

Sato, 2007, Nitric Oxide Plays a Critical Role in Suppression of T-cell Proliferation by Mesenchymal Stem Cells, Blood, 109, 228, 10.1182/blood-2006-02-002246

Ge, 2019, Mechanisms of the Immunomodulation Effects of Bone Marrow-Derived Mesenchymal Stem Cells on Facial Nerve Injury in Sprague-Dawley Rats, Stem Cells Dev., 28, 489, 10.1089/scd.2018.0104

Chen, 2020, Mesenchymal Stem Cells Regulate the Th17/Treg Cell Balance Partly through Hepatocyte Growth Factor In Vitro, Stem Cell Res. Ther., 11, 91, 10.1186/s13287-020-01612-y

Cao, 2016, Hepatocyte Growth Factor Enhances the Inflammation-Alleviating Effect of Umbilical Cord-Derived Mesenchymal Stromal Cells in a Bronchiolitis Obliterans Model, Cytotherapy, 18, 402, 10.1016/j.jcyt.2015.12.006

Hieronymus, 2015, HGF/Met-Signaling Contributes to Immune Regulation by Modulating Tolerogenic and Motogenic Properties of Dendritic Cells, Biomedicines, 3, 138, 10.3390/biomedicines3010138

Keir, 2008, PD-1 and its ligands in tolerance and immunity, Annu. Rev. Immunol., 26, 677, 10.1146/annurev.immunol.26.021607.090331

Kim, 2018, Tonsil-derived mesenchymal stem cells (T-MSCs) prevent Th17-mediated autoimmune response via regulation of the programmed death-1/programmed death ligand-1 (PD-1/PD-L1) pathway, J. Tissue Eng. Regen. Med., 12, e1022, 10.1002/term.2423

Djouad, 2016, Mesenchymal Stem Cell-Derived Interleukin 1 Receptor Antagonist Promotes Macrophage Polarization and Inhibits B Cell Differentiation, Stem Cells, 34, 483, 10.1002/stem.2254

Ren, 2010, Inflammatory Cytokine-Induced Intercellular Adhesion Molecule-1 and Vascular Cell Adhesion Molecule-1 in Mesenchymal Stem Cells Are Critical for Immunosuppression, J. Immunol., 184, 2321, 10.4049/jimmunol.0902023

Colombo, 2014, Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles, Annu. Rev. Cell Dev. Biol., 30, 255, 10.1146/annurev-cellbio-101512-122326

Preciado, 2016, MSC Surface Markers (CD44, CD73, and CD90) Can Identify Human MSC-derived Extracellular Vesicles by Conventional Flow Cytometry, Cell Commun. Signal., 14, 2, 10.1186/s12964-015-0124-8

Quesenberry, 2014, Cellular Phenotype and Extracellular Vesicles: Basic and Clinical Considerations, Stem Cells Dev., 23, 1429, 10.1089/scd.2013.0594

Montecalvo, 2012, Mechanism of Transfer of Functional microRNAs between Mouse Dendritic Cells via Exosomes, Blood, 119, 756, 10.1182/blood-2011-02-338004

Galvez, 2018, Can a Conversation between Mesenchymal Stromal Cells and Macrophages Solve the Crisis in the Inflamed Intestine?, Front. Pharmacol., 9, 179, 10.3389/fphar.2018.00179

Rad, 2019, Mesenchymal Stem Cell-Based Therapy for Autoimmune Diseases: Emerging Roles of Extracellular Vesicles, Mol. Biol. Rep., 46, 1533, 10.1007/s11033-019-04588-y

Zheng, 2019, Exosomal microRNA-155-5p from PDLSCs Regulated Th17/Treg Balance by Targeting Sirtuin-1 in Chronic Periodontitis, J. Cell. Physiol., 234, 20662, 10.1002/jcp.28671

Song, 2018, miR-21 Protects against Ischemia/Reperfusion-Induced Acute Kidney Injury by Preventing Epithelial Cell Apoptosis and Inhibiting Dendritic Cell Maturation, Front. Physiol., 9, 790, 10.3389/fphys.2018.00790

Gu, 2016, Mesenchymal Stromal Cells Derived Extracellular Vesicles Ameliorate Acute Renal Ischemia Reperfusion Injury by Inhibition of Mitochondrial Fission through miR-30, Stem Cells Int., 2016, 2093940, 10.1155/2016/2093940

Harrell, C.R., Fellabaum, C., Jovicic, N., Djonov, V., Arsenijevic, N., and Volarevic, V. (2019). Molecular Mechanisms Responsible for Therapeutic Potential of Mesenchymal Stem Cell-Derived Secretome. Cells, 8.

Kapnick, 2018, The Emerging Role of Immune Dysfunction in Mitochondrial Diseases as a Paradigm for Understanding Immunometabolism, Metab. Clin. Exp., 81, 97, 10.1016/j.metabol.2017.11.010

Torralba, 2016, Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer, Front. Cell Dev. Biol., 4, 107, 10.3389/fcell.2016.00107

Berridge, 2016, Horizontal Transfer of Mitochondria between Mammalian Cells: Beyond Co-Culture Approaches, Curr. Opin. Genet. Dev., 38, 75, 10.1016/j.gde.2016.04.003

Fan, 2020, Mechanisms Underlying the Protective Effects of Mesenchymal Stem Cell-Based Therapy, Cell. Mol. Life Sci., 77, 2771, 10.1007/s00018-020-03454-6

Wang, 2018, Cell Adhesion-Mediated Mitochondria Transfer Contributes to Mesenchymal Stem Cell-Induced Chemoresistance on T Cell Acute Lymphoblastic Leukemia Cells, J. Hematol. Oncol., 11, 11, 10.1186/s13045-018-0554-z

Ahmad, 2014, Miro1 Regulates Intercellular Mitochondrial Transport & Enhances Mesenchymal Stem Cell Rescue Efficacy, EMBO J., 33, 994

Kim, 2018, Delivery of Exogenous Mitochondria via Centrifugation Enhances Cellular Metabolic Function, Sci. Rep., 8, 3330, 10.1038/s41598-018-21539-y

Konari, 2019, Mitochondria Transfer from Mesenchymal Stem Cells Structurally and Functionally Repairs Renal Proximal Tubular Epithelial Cells in Diabetic Nephropathy In Vivo, Sci. Rep., 9, 5184, 10.1038/s41598-019-40163-y

Islam, 2012, Mitochondrial Transfer from Bone-Marrow-Derived Stromal Cells to Pulmonary Alveoli Protects against Acute Lung Injury, Nat. Med., 18, 759, 10.1038/nm.2736

Jiang, 2016, Mitochondrial Transfer of Mesenchymal Stem Cells Effectively Protects Corneal Epithelial Cells From Mitochondrial Damage, Cell Death Dis., 7, e2467, 10.1038/cddis.2016.358

Torres, 2016, The Immunosuppressive Signature of Menstrual Blood Mesenchymal Stem Cells Entails Opposite Effects on Experimental Arthritis and Graft versus Host Diseases, Stem Cells, 34, 456, 10.1002/stem.2244

Noronha, 2019, Priming Approaches to Improve the Efficacy of Mesenchymal Stromal Cell-Based Therapies, Stem Cell Res. Ther., 10, 131, 10.1186/s13287-019-1224-y

Wang, 2016, Comparative Analysis of Human Mesenchymal Stem Cells from Fetal-Bone Marrow, Adipose Tissue, and Warton’s Jelly as Sources of Cell Immunomodulatory Therapy, Hum. Vaccines Immunother., 12, 85, 10.1080/21645515.2015.1030549

Carrero, 2012, IL1β Induces Mesenchymal Stem Cells Migration and Leucocyte Chemotaxis Through NF-κB, Stem Cell Rev. Rep., 8, 905, 10.1007/s12015-012-9364-9

Fan, 2012, Pre-treatment with IL-1β Enhances the Efficacy of MSC Transplantation in DSS-induced Colitis, Cell. Mol. Immunol., 9, 473, 10.1038/cmi.2012.40

Han, 2014, Interleukin-17 Enhances Immunosuppression by Mesenchymal Stem Cells, Cell Death Differ., 21, 1758, 10.1038/cdd.2014.85

Kurte, 2018, IL17/IL17RA As a Novel Signaling Axis Driving Mesenchymal Stem Cell Therapeutic Function in Experimental Autoimmune Encephalomyelitis, Front. Immunol., 9, 802, 10.3389/fimmu.2018.00802

Sivanathan, 2015, Interleukin-17A-Induced Human Mesenchymal Stem Cells Are Superior Modulators of Immunological Function, Stem Cells, 33, 2850, 10.1002/stem.2075

Zimmermann, 2014, Pre-Conditioning Mesenchymal Stromal Cell Spheroids for Immunomodulatory Paracrine Factor Secretion, Cytotherapy, 16, 331, 10.1016/j.jcyt.2013.09.004

Spencer, 2014, Direct Measurement of Local Oxygen Concentration in the Bone Marrow of Live Animals, Nature, 508, 269, 10.1038/nature13034

Saparov, 2016, Preconditioning of Human Mesenchymal Stem Cells to Enhance Their Regulation of the Immune Response, Stem Cells Int., 2016, 3924858, 10.1155/2016/3924858

Haque, 2013, Hypoxic Culture Conditions As a Solution for Mesenchymal Stem Cell Based Regenerative Therapy, Sci. World J., 2013, 632972, 10.1155/2013/632972

Paredes, 2020, HIF1α-Dependent Metabolic Reprogramming Governs Mesenchymal Stem/Stromal Cell Immunoregulatory Functions, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol., 34, 8250

Grimaldi, 2010, Metabolic and Nonmetabolic Regulatory Functions of Peroxisome Proliferator-Activated Receptor Beta, Curr. Opin. Lipidol., 21, 186, 10.1097/MOL.0b013e32833884a4

Palomer, X., Barroso, E., Pizarro-Delgado, J., Peña, L., Botteri, G., Zarei, M., Aguilar, D., Montori-Grau, M., and Vázquez-Carrera, M. (2018). PPARβ/δ: A Key Therapeutic Target in Metabolic Disorders. Int. J. Mol. Sci., 19.

Ipseiz, 2016, PPARβ/δ Directs the Therapeutic Potential of Mesenchymal Stem Cells in Arthritis, Ann. Rheum. Dis., 75, 2166, 10.1136/annrheumdis-2015-208696

Ipseiz, 2018, Gilz-Activin A As a Novel Signaling Axis Orchestrating Mesenchymal Stem Cell and Th17 Cell Interplay, Theranostics, 8, 846, 10.7150/thno.21793

Palkar, 2010, Cellular and Pharmacological Selectivity of the Peroxisome Proliferator-Activated Receptor-β/δ Antagonist GSK3787, Mol. Pharmacol., 78, 419, 10.1124/mol.110.065508

Hong, F., Xu, P., and Zhai, Y. (2018). The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci., 19.