Involvement of Mesenchymal Stem Cells in Oral Mucosal Bacterial Immunotherapy

Alberto Vázquez, Lidia M Fernández-Sevilla, Eva Jiménez, David Pérez-Cabrera, Rosa Yañez, Jose Luis Subiza, Alberto Varas, Jaris Valencia, Angeles Vicente, Alberto Vázquez, Lidia M Fernández-Sevilla, Eva Jiménez, David Pérez-Cabrera, Rosa Yañez, Jose Luis Subiza, Alberto Varas, Jaris Valencia, Angeles Vicente

Abstract

Recent clinical observations indicate that bacterial vaccines induce cross-protection against infections produced by different microorganisms. MV130, a polyvalent bacterial sublingual preparation designed to prevent recurrent respiratory infectious diseases, reduces the infection rate in patients with recurrent respiratory tract infections. On the other hand, mesenchymal stem cells (MSCs) are key cell components that contribute to the maintenance of tissue homeostasis and exert both immunostimulatory and immunosuppressive functions. Herein, we study the effects of MV130 in human MSC functionality as a potential mechanism that contributes to its clinical benefits. We provide evidence that during MV130 sublingual immunization of mice, resident oral mucosa MSCs can take up MV130 components and their numbers remain unchanged after vaccination, in contrast to granulocytes that are recruited from extramucosal tissues. MSCs treated in vitro with MV130 show an increased viability without affecting their differentiation potential. In the short-term, MSC treatment with MV130 induces higher leukocyte recruitment and T cell expansion. In contrast, once T-cell activation is initiated, MV130 stimulation induces an up-regulated expression of immunosuppressor factors in MSCs. Accordingly, MV130-primed MSCs reduce T lymphocyte proliferation, induce the differentiation of dendritic cells with immunosuppressive features and favor M2-like macrophage polarization, thus counterbalancing the immune response. In addition, MSCs trained with MV130 undergo functional changes, enhancing their immunomodulatory response to a secondary stimulus. Finally, we show that MSCs are able to uptake, process and retain a reservoir of the TLR ligands derived from MV130 digestion which can be subsequently transferred to dendritic cells, an additional feature that also may be associated to trained immunity.

Keywords: immunomodulation; mesenchymal stem cells; pattern recognition receptors; polybacterial preparation; short-term memory; sublingual mucosal immunotherapy; vaccine.

Conflict of interest statement

JS is the CEO of Inmunotek SL, a pharmaceutical company that manufactures bacterial vaccines. The authors declare that this study received funding from Inmunotek SL. The funder had the following involvement in the study: final writing and editing of the manuscript.

Copyright © 2020 Vázquez, Fernández-Sevilla, Jiménez, Pérez-Cabrera, Yañez, Subiza, Varas, Valencia and Vicente.

Figures

Scheme 1
Scheme 1
Figure 1
Figure 1
Treatment with MV130 modifies biological properties of MSCs. MSCs were treated with MV130 for 2 h. (A, B) After 24 h, MSCs were stained with Annexin V and IP and cell viability was analysed by flow cytometry. Mean ± SEM of 5 independent experiments (A) and a representative experiment (B) are shown. (C) MSCs were treated with MV130 for 2 h. Bcl2 and Bcl-xL expression in MSCs were analyzed by flow cytometry 24 h after treatment. The percentages of positive cells are indicated in each histogram. Gray filled histograms represent isotype control staining. Data are representative of 4 independent experiments. (D) 48 h after treatment, the expression of different surface markers was studied on MSCs by flow cytometry. Representative histograms and MFI values are shown (n = 3-4). Gray histograms represent isotype controls. (E) mRNA expression for different immunomodulatory factors was studied on MSCs by qRT-PCR. Data represent mean ± SEM of 8 to 12 independent experiments relative to individual controls. (F) Supernatants from MSC cultures were collected 48 h after treatment. Protein secretion relative to individual controls is expressed as mean ± SEM from 15 independent experiments (*p < 0.05; **p < 0.01***p < 0.005 by Wilcoxon test).
Figure 2
Figure 2
MV130 is uptaken by oral MSCs in vivo. (A) Flow cytometry analysis of T lymphocytes (CD45+CD3+), macrophages (F4/80+MHC-IIlo), DCs (F4/80-MHC-II+) and MSCs (CD29+Sca-1+) present in the oral mucosa from mice after sublingual immunization with MV130-CFSE. Histograms show the percentage of uptake of CFSE-MV130 by each of these populations. Data are representative of 3 independent experiments. (B) Percentage of T cells, granulocytes and MSCs present in peripheral lymph nodes (P-LN), submaxillary lymph nodes (SM-LN) and oral mucosa (OM) from mice after sublingual immunization with MV130 respect to control mice (n = 6–13). (C) MV130 priming reduces MSC migration capacity. Bar graph shows the percentage of migrating cells in MV130 primed cultures. Results represent the mean ± SEM of 3 independent experiments (D) MV130 does not specifically attract MSCs. Bar graph shows the percentage of migrating cells to MV130 or IFNγ. Results represent the mean ± SEM of 4 independent experiments. (*p < 0.05; ***p < 0.005 by Wilcoxon test).
Figure 3
Figure 3
MSCs act as reservoirs of MV130 and are able to transfer it to DCs. MSCs were treated for 24 h with MV130 labeled with CFSE (CFSE-MV130; green) for monitoring. After washing cells to remove the drug, its uptake, processing and transference to DCs were studied. (A) Uptake and maintenance of CFSE-MV130. Histograms show the percentage of positive cells and MFI in brackets (n = 6–8 independent experiments). (B, C) Spatiotemporal monitoring of MV130 (green) in MSCs. Immunostaining in red for CD63 (B) or LAMP2 (C) in MV130-MSCs at different times. Inset indicated by arrows in B, scale bar: 10μm (24 h) or 1μm (120 h). Right: Higher magnification of the white square in (C) (120 h); examples of LAMP2-positive compartments with CFSE-MV130. Hoechst was used for nucleus staining (blue). Images are representative of 5 independent experiments. (D–G) DCs differentiated from monocytes were co-cultured with MSCs, previously treated with MV130 or CFSE-MV130 as described in Material & Methods section, and transfer of MV130 from MSCs to DCs was studied. A representative dot plot (D) and the mean ± SEM of five independent experiments (E) are shown. DCs and MSCs were gated according to CD1a or CD90 expression, respectively. In (E) control MSCs and DCs directly treated with CFSE-MV130 are also shown. (F) MV130 transfer from MSCs to DCs studied by immunofluorescence. Hoechst was used for nucleus staining in all cases (blue). Co-cultures were labeled with phalloidin (red) and anti-HLA-DR (magenta). The absence of HLA-DR expression on MSCs allow to distinguish it from DCs (HLA-DR+). White arrows in (F) indicate area of insert image magnification. (G) MSCs were labeled with anti-CD63 (red) and Hoechst was used for nucleus staining (blue). Co-localization of CD63+ extracellular microvesicles with CFSE-MV130 was observed free in the medium. White arrows indicate area of insert image magnification. Representative images of 3 independent experiments (*p < 0.05 significance by Wilcoxon test).
Figure 4
Figure 4
Immunomodulatory abilities of MSCs after activation with MV130. (A–D) Phenotype and function of monocyte-derived DCs differentiated in the presence or absence of CTRL-MSCs or MV130-MSCs. At day 6, CD1a and CD14 expression were analyzed by flow cytometry in the CD90- population. The percentage of positive cells is shown in each plot (A) and IL-6 production was measured in the supernatants (B). Results represent the mean ± SEM (n = 6). (C, D) DCs stimulated with LPS were cultured in MLR assays with CFSE-labeled T lymphocytes. After 5 days, the percentage of proliferating T cells was calculated by CFSE dilution method (gated on CD3+ cell population) (C) and supernatants from MLR co-cultures were analyzed for TNFα and IL-10 protein secretion (D). Data represent the mean ± SEM (n = 3–5). (E) Control or MV130-MSCs were co-cultured with CFSE-labeled T lymphocytes stimulated with CD3/CD28 beads, for different times. Histograms show CFSE staining in proliferating T cells in CD3+ gated cells. Proliferation index referred to unstimulated T lymphocytes (gray line) is indicated. Data are representative from four independent experiments. (F) MV130-MSCs re-stimulated with IFNγ, following protocol described in Material and Methods, were co-cultured with CFSE labeled T lymphocytes. Proliferation index is shown. Bar graph shows mean ± SEM (n = 4). (G–K) Control and MV130 primed MSCs were co-cultured with monocytes in the presence of GM-CSF to induce M1 macrophage differentiation. Monocytes alone were cultured as M1 control. (G–I) After 6 days, CD14, CD163 and PD-L2 expression was determined by flow cytometry in non-MSC population (CD90- cells) (n = 5–6). A representative experiment (G) and mean ± SEM of percentage of CD14+CD163+CD90- cells from five to six independent experiments (H) are shown. (I) Representative PD-L2 expression on macrophages. MFI is shown in each histogram. (J) After 6 days of co-culture, LPS was added and supernatants were analysed for TNFα and IL-10 production. Data represent TNFα/IL-10 ratio production at the different experimental conditions (mean ± SEM; n = 4). (K) Macrophages stimulated with LPS were used to carry out MLR cultures with CFSE-labeled T lymphocytes. After 5 days, the percentage of T cell proliferation was measured in the CD3+ cell population. (L) CCL2 and CXCL8 protein secretion measured in control and MV130-MSC culture supernatants. Bars represent the mean ± SEM relative to individual controls from 15 independent experiments. (M) PBMCs were placed in a transwell insert while MSCs, treated with or without MV130 for the 24 h previous, and seeded in the bottom chamber. After 8 h, migrating PBMCs (present in the lower chamber) were collected and stained for CD14, CD56, CD3, HLA-DR, and CD19, and different leukocyte populations were analyzed by flow cytometry. MSCs were excluded from the analysis by CD90 expression. (mean ± SEM; n = 4) (N) PBMCs migrating toward control or MV130 primed MSCs re-stimulated with IFNγ. Monocyte recruitment was analyzed by flow cytometry (mean ± SEM, n = 4). (O) Supernatants from MSC cultures following the protocol described in Material & Methods section were analyzed for CCL2, CXCL8, and CXCL10 protein secretion after IFNγ re-stimulation. Results represent mean ± SEM of four to six independent experiments relative to individual controls. (*p < 0.05; **p < 0.01, ***p < 0.005 significances relative to M1-macrophages or DC; #p < 0.05; ###p < 0.005 significances relative to CTRL-MSCs by Wilcoxon test).
Figure 5
Figure 5
Effects of MV130-MSCs in an in vivo model of acute inflammation. FVB/NJ mice were challenged in the footpad with 40μg of LPS and administered with or without control or MV130-primed MSCs 24 h later. (A) Footpad thickness increment was determined after 72 h as a measure of the efficacy of the different experimental groups of MSCs. Data shown are mean ± SEM of two independent experiments (three mice per group) (B) Images show histological sections of footpad tissue stained with Gallego’s Trichrome. Images are representative of 3 mice per group. (C) Percentage of CD45+ leukocytes infiltrating footpads in the different mouse groups analyzed by flow cytometry. The distribution of the different leukocyte subpopulations in CD45+ cells is also shown in each experimental group. Results represent increments relative to control animals (2 independent experiments with 3 mice per group) (*p < 0.05,**p < 0.01 versus LPS alone; #p < 0.05 versus CTRL-MSCs; by Wilcoxon test).
Figure 6
Figure 6
Proposed Model of MSC involvement in oral mucosal bacterial immunotherapy with MV130.

References

    1. Guthrie R. Community-acquired lower respiratory tract infections: etiology and treatment. Chest (2001) 120(6):2021–34. 10.1378/chest.120.6.2021
    1. Marengo R, Ortega Martell JA, Esposito S. Paediatric Recurrent Ear, Nose and Throat Infections and Complications: Can We Do More? Infect Dis Ther (2020) 9(2):275–90. 10.1007/s40121-020-00289-3
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PloS Biol (2008) 6(11):e280. 10.1371/journal.pbio.0060280
    1. Esposito S, Daleno C, Tagliabue C, Scala A, Tenconi R, Borzani I, et al. Impact of rhinoviruses on pediatric community-acquired pneumonia. Eur J Clin Microbiol Infect Dis (2012) 31(7):1637–45. 10.1007/s10096-011-1487-4
    1. Blaser MJ. Antibiotic use and its consequences for the normal microbiome. Science (2016) 352(6285):544–5. 10.1126/science.aad9358
    1. Godman B, Haque M, McKimm J, Abu Bakar M, Sneddon J, Wale J, et al. Ongoing strategies to improve the management of upper respiratory tract infections and reduce inappropriate antibiotic use particularly among lower and middle-income countries: findings and implications for the future. Curr Med Res Opin (2020) 36(2):301–27. 10.1080/03007995.2019.1700947
    1. Tejera-Alhambra M, Palomares O, Perez de Diego R, Diaz-Lezcano I, Sanchez-Ramon S. New Biological Insights in the Immunomodulatory Effects of Mucosal Polybacterial Vaccines in Clinical Practice. Curr Pharm Des (2016) 22(41):6283–93. 10.2174/1381612822666160829143129
    1. Steurer-Stey C, Bachmann LM, Steurer J, Tramer MR. Oral purified bacterial extracts in chronic bronchitis and COPD: systematic review. Chest (2004) 126(5):1645–55. 10.1378/chest.126.5.1645
    1. Alecsandru D, Valor L, Sanchez-Ramon S, Gil J, Carbone J, Navarro J, et al. Sublingual therapeutic immunization with a polyvalent bacterial preparation in patients with recurrent respiratory infections: immunomodulatory effect on antigen-specific memory CD4+ T cells and impact on clinical outcome. Clin Exp Immunol (2011) 164(1):100–7. 10.1111/j.1365-2249.2011.04320.x
    1. Pasquali C, Salami O, Taneja M, Gollwitzer ES, Trompette A, Pattaroni C, et al. Enhanced Mucosal Antibody Production and Protection against Respiratory Infections Following an Orally Administered Bacterial Extract. Front Med (Lausanne) (2014) 1:41. 10.3389/fmed.2014.00041
    1. Garcia Gonzalez LA, Arrutia Diez F. Mucosal bacterial immunotherapy with MV130 highly reduces the need of tonsillectomy in adults with recurrent tonsillitis. Hum Vaccin Immunother (2019) 15(9):2150–3. 10.1080/21645515.2019.1581537
    1. Negri DR, Riccomi A, Pinto D, Vendetti S, Rossi A, Cicconi R, et al. Persistence of mucosal and systemic immune responses following sublingual immunization. Vaccine (2010) 28(25):4175–80. 10.1016/j.vaccine.2010.04.013
    1. Benito-Villalvilla C, Cirauqui C, Diez-Rivero CM, Casanovas M, Subiza JL, Palomares O. MV140, a sublingual polyvalent bacterial preparation to treat recurrent urinary tract infections, licenses human dendritic cells for generating Th1, Th17, and IL-10 responses via Syk and MyD88. Mucosal Immunol (2017) 10(4):924–35. 10.1038/mi.2016.112
    1. Sanchez-Ramon S, Conejero L, Netea MG, Sancho D, Palomares O, Subiza JL. Trained Immunity-Based Vaccines: A New Paradigm for the Development of Broad-Spectrum Anti-infectious Formulations. Front Immunol (2018) 9:2936. 10.3389/fimmu.2018.02936
    1. Parola C, Salogni L, Vaira X, Scutera S, Somma P, Salvi V, et al. Selective activation of human dendritic cells by OM-85 through a NF-kB and MAPK dependent pathway. PloS One (2013) 8(12):e82867. 10.1371/journal.pone.0082867
    1. Dang AT, Pasquali C, Ludigs K, Guarda G. OM-85 is an immunomodulator of interferon-beta production and inflammasome activity. Sci Rep (2017) 7:43844. 10.1038/srep43844
    1. Cirauqui C, Benito-Villalvilla C, Sanchez-Ramon S, Sirvent S, Diez-Rivero CM, Conejero L, et al. Human dendritic cells activated with MV130 induce Th1, Th17 and IL-10 responses via RIPK2 and MyD88 signalling pathways. Eur J Immunol (2018) 48(1):180–93. 10.1002/eji.201747024
    1. Hamada A, Torre C, Drancourt M, Ghigo E. Trained Immunity Carried by Non-immune Cells. Front Microbiol (2018) 9:3225. 10.3389/fmicb.2018.03225
    1. Molero-Abraham M, Sanchez-Trincado JL, Gomez-Perosanz M, Torres-Gomez A, Subiza JL, Lafuente EM, et al. Human Oral Epithelial Cells Impair Bacteria-Mediated Maturation of Dendritic Cells and Render T Cells Unresponsive to Stimulation. Front Immunol (2019) 10:1434. 10.3389/fimmu.2019.01434
    1. Zhou LL, Liu W, Wu YM, Sun WL, Dorfer CE, Fawzy El-Sayed KM. Oral Mesenchymal Stem/Progenitor Cells: The Immunomodulatory Masters. Stem Cells Int (2020) 2020:1327405. 10.1155/2020/1327405
    1. Marynka-Kalmani K, Treves S, Yafee M, Rachima H, Gafni Y, Cohen MA, et al. The lamina propria of adult human oral mucosa harbors a novel stem cell population. Stem Cells (2010) 28(5):984–95. 10.1002/stem.425
    1. Togarrati PP, Sasaki RT, Abdel-Mohsen M, Dinglasan N, Deng X, Desai S, et al. Identification and characterization of a rich population of CD34(+) mesenchymal stem/stromal cells in human parotid, sublingual and submandibular glands. Sci Rep (2017) 7(1):3484. 10.1038/s41598-017-03681-1
    1. Togarrati PP, Dinglasan N, Desai S, Ryan WR, Muench MO. CD29 is highly expressed on epithelial, myoepithelial, and mesenchymal stromal cells of human salivary glands. Oral Dis (2018) 24(4):561–72. 10.1111/odi.12812
    1. Shirjang S, Mansoori B, Solali S, Hagh MF, Shamsasenjan K. Toll-like receptors as a key regulator of mesenchymal stem cell function: An up-to-date review. Cell Immunol (2017) 315:1–10. 10.1016/j.cellimm.2016.12.005
    1. Meisel R, Brockers S, Heseler K, Degistirici O, Bulle H, Woite C, et al. Human but not murine multipotent mesenchymal stromal cells exhibit broad-spectrum antimicrobial effector function mediated by indoleamine 2,3-dioxygenase. Leukemia (2011) 25(4):648–54. 10.1038/leu.2010.310
    1. Sung DK, Chang YS, Sung SI, Yoo HS, Ahn SY, Park WS. Antibacterial effect of mesenchymal stem cells against Escherichia coli is mediated by secretion of beta- defensin- 2 via toll- like receptor 4 signalling. Cell Microbiol (2016) 18(3):424–36. 10.1111/cmi.12522
    1. Alcayaga-Miranda F, Cuenca J, Khoury M. Antimicrobial Activity of Mesenchymal Stem Cells: Current Status and New Perspectives of Antimicrobial Peptide-Based Therapies. Front Immunol (2017) 8:339. 10.3389/fimmu.2017.00339
    1. Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell (2013) 13(4):392–402. 10.1016/j.stem.2013.09.006
    1. Brandau S, Jakob M, Hemeda H, Bruderek K, Janeschik S, Bootz F, et al. Tissue-resident mesenchymal stem cells attract peripheral blood neutrophils and enhance their inflammatory activity in response to microbial challenge. J Leukoc Biol (2010) 88(5):1005–15. 10.1189/jlb.0410207
    1. Weiss ARR, Dahlke MH. Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front Immunol (2019) 10:1191. 10.3389/fimmu.2019.01191
    1. Waterman RS, Tomchuck SL, Henkle SL, Betancourt AM. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PloS One (2010) 5(4):e10088. 10.1371/journal.pone.0010088
    1. Dumitru CA, Hemeda H, Jakob M, Lang S, Brandau S. Stimulation of mesenchymal stromal cells (MSCs) via TLR3 reveals a novel mechanism of autocrine priming. FASEB J (2014) 28(9):3856–66. 10.1096/fj.14-250159
    1. Najar M, Krayem M, Meuleman N, Bron D, Lagneaux L. Mesenchymal Stromal Cells and Toll-Like Receptor Priming: A Critical Review. Immune Netw (2017) 17(2):89–102. 10.4110/in.2017.17.2.89
    1. Martinez VG, Ontoria-Oviedo I, Ricardo CP, Harding SE, Sacedon R, Varas A, et al. Overexpression of hypoxia-inducible factor 1 alpha improves immunomodulation by dental mesenchymal stem cells. Stem Cell Res Ther (2017) 8(1):208. 10.1186/s13287-017-0659-2
    1. Mekhemar MK, Adam-Klages S, Kabelitz D, Dorfer CE, Fawzy El-Sayed KM. TLR-induced immunomodulatory cytokine expression by human gingival stem/progenitor cells. Cell Immunol (2018) 326:60–7. 10.1016/j.cellimm.2017.01.007
    1. Liu GY, Liu Y, Lu Y, Qin YR, Di GH, Lei YH, et al. Short-term memory of danger signals or environmental stimuli in mesenchymal stem cells: implications for therapeutic potential. Cell Mol Immunol (2016) 13(3):369–78. 10.1038/cmi.2015.11
    1. Lin T, Pajarinen J, Kohno Y, Huang JF, Maruyama M, Romero-Lopez M, et al. Trained murine mesenchymal stem cells have anti-inflammatory effect on macrophages, but defective regulation on T-cell proliferation. FASEB J (2019) 33(3):4203–11. 10.1096/fj.201801845R
    1. Lopez-Santalla M, Hervas-Salcedo R, Fernandez-Garcia M, Bueren JA, Garin MI. Cell therapy with mesenchymal stem cells induces an innate immune memory response that attenuates experimental colitis in the long-term. J Crohns Colitis (2020) 14(10):1424–35. 10.1093/ecco-jcc/jjaa079
    1. Gonzalez-Rey E, Anderson P, Gonzalez MA, Rico L, Buscher D, Delgado M. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut (2009) 58(7):929–39. 10.1136/gut.2008.168534
    1. Krasnodembskaya A, Song Y, Fang X, Gupta N, Serikov V, Lee JW, et al. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells (2010) 28(12):2229–38. 10.1002/stem.544
    1. Gupta N, Krasnodembskaya A, Kapetanaki M, Mouded M, Tan X, Serikov V, et al. Mesenchymal stem cells enhance survival and bacterial clearance in murine Escherichia coli pneumonia. Thorax (2012) 67(6):533–9. 10.1136/thoraxjnl-2011-201176
    1. Lee JW, Krasnodembskaya A, McKenna DH, Song Y, Abbott J, Matthay MA. Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria. Am J Respir Crit Care Med (2013) 187(7):751–60. 10.1164/rccm.201206-0990OC
    1. Park J, Kim S, Lim H, Liu A, Hu S, Lee J, et al. Therapeutic effects of human mesenchymal stem cell microvesicles in an ex vivo perfused human lung injured with severe E. coli pneumonia. Thorax (2019) 74(1):43–50. 10.1136/thoraxjnl-2018-211576
    1. Vicente Lopez MA, Vazquez Garcia MN, Entrena A, Olmedillas Lopez S, Garcia-Arranz M, Garcia-Olmo D, et al. Low doses of bone morphogenetic protein 4 increase the survival of human adipose-derived stem cells maintaining their stemness and multipotency. Stem Cells Dev (2011) 20(6):1011–9. 10.1089/scd.2010.0355
    1. Hammond MD, Ai Y, Sansing LH. Gr1+ Macrophages and Dendritic Cells Dominate the Inflammatory Infiltrate 12 Hours After Experimental Intracerebral Hemorrhage. Transl Stroke Res (2012) 3(1):s125–s31. 10.1007/s12975-012-0174-9
    1. Fraile-Ramos A, Kledal TN, Pelchen-Matthews A, Bowers K, Schwartz TW, Marsh M. The human cytomegalovirus US28 protein is located in endocytic vesicles and undergoes constitutive endocytosis and recycling. Mol Biol Cell (2001) 12(6):1737–49. 10.1091/mbc.12.6.1737
    1. Weinstock A, Pevsner-Fischer M, Porat Z, Selitrennik M, Zipori D. Cultured Mesenchymal Stem Cells Stimulate an Immune Response by Providing Immune Cells with Toll-Like Receptor 2 Ligand. Stem Cell Rev Rep (2015) 11(6):826–40. 10.1007/s12015-015-9614-8
    1. Rafei M, Campeau PM, Aguilar-Mahecha A, Buchanan M, Williams P, Birman E, et al. Mesenchymal stromal cells ameliorate experimental autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC chemokine ligand 2-dependent manner. J Immunol (2009) 182(10):5994–6002. 10.4049/jimmunol.0803962
    1. Gschwandtner M, Derler R, Midwood KS. More Than Just Attractive: How CCL2 Influences Myeloid Cell Behavior Beyond Chemotaxis. Front Immunol (2019) 10:2759. 10.3389/fimmu.2019.02759
    1. Giri J, Das R, Nylen E, Chinnadurai R, Galipeau J. CCL2 and CXCL12 Derived from Mesenchymal Stromal Cells Cooperatively Polarize IL-10+ Tissue Macrophages to Mitigate Gut Injury. Cell Rep (2020) 30(6):1923–34 e4. 10.1016/j.celrep.2020.01.047
    1. Moutsopoulos NM, Moutsopoulos HM. The oral mucosa: A barrier site participating in tissue-specific and systemic immunity. Oral Dis (2018) 24(1-2):22–5. 10.1111/odi.12729
    1. Iwamura C, Bouladoux N, Belkaid Y, Sher A, Jankovic D. Sensing of the microbiota by NOD1 in mesenchymal stromal cells regulates murine hematopoiesis. Blood (2017) 129(2):171–6. 10.1182/blood-2016-06-723742
    1. Delarosa O, Dalemans W, Lombardo E. Toll-like receptors as modulators of mesenchymal stem cells. Front Immunol (2012) 3:182. 10.3389/fimmu.2012.00182
    1. Lombardo E, DelaRosa O, Mancheno-Corvo P, Menta R, Ramirez C, Buscher D. Toll-like receptor-mediated signaling in human adipose-derived stem cells: implications for immunogenicity and immunosuppressive potential. Tissue Eng Part A (2009) 15(7):1579–89. 10.1089/ten.tea.2008.0340
    1. Nagai Y, Shiraishi D, Tanaka Y, Nagasawa Y, Ohwada S, Shimauchi H, et al. Transportation of sublingual antigens across sublingual ductal epithelial cells to the ductal antigen-presenting cells in mice. Clin Exp Allergy (2015) 45(3):677–86. 10.1111/cea.12329
    1. Song JH, Kim JI, Kwon HJ, Shim DH, Parajuli N, Cuburu N, et al. CCR7-CCL19/CCL21-regulated dendritic cells are responsible for effectiveness of sublingual vaccination. J Immunol (2009) 182(11):6851–60. 10.4049/jimmunol.0803568
    1. Lei J, Wang Z, Hui D, Yu W, Zhou D, Xia W, et al. Ligation of TLR2 and TLR4 on murine bone marrow-derived mesenchymal stem cells triggers differential effects on their immunosuppressive activity. Cell Immunol (2011) 271(1):147–56. 10.1016/j.cellimm.2011.06.014
    1. Kalimuthu S, Oh JM, Gangadaran P, Zhu L, Lee HW, Rajendran RL, et al. In Vivo Tracking of Chemokine Receptor CXCR4-Engineered Mesenchymal Stem Cell Migration by Optical Molecular Imaging. Stem Cells Int (2017) 2017:8085637. 10.1155/2017/8085637
    1. van Megen KM, van ‘t Wout ET, Lages Motta J, Dekker B, Nikolic T, Roep BO. Activated Mesenchymal Stromal Cells Process and Present Antigens Regulating Adaptive Immunity. Front Immunol (2019) 10:694. 10.3389/fimmu.2019.00694
    1. Wang Y, Chen X, Cao W, Shi Y. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol (2014) 15(11):1009–16. 10.1038/ni.3002
    1. Najar M, Krayem M, Merimi M, Burny A, Meuleman N, Bron D, et al. Insights into inflammatory priming of mesenchymal stromal cells: functional biological impacts. Inflammation Res (2018) 67(6):467–77. 10.1007/s00011-018-1131-1
    1. Kota DJ, DiCarlo B, Hetz RA, Smith P, Cox CS, Jr., Olson SD. Differential MSC activation leads to distinct mononuclear leukocyte binding mechanisms. Sci Rep (2014) 4:4565. 10.1038/srep04565
    1. Krampera M. Mesenchymal stromal cell ‘licensing’: a multistep process. Leukemia (2011) 25(9):1408–14. 10.1038/leu.2011.108
    1. Duijvestein M, Wildenberg ME, Welling MM, Hennink S, Molendijk I, van Zuylen VL, et al. Pretreatment with interferon-gamma enhances the therapeutic activity of mesenchymal stromal cells in animal models of colitis. Stem Cells (2011) 29(10):1549–58. 10.1002/stem.698
    1. Liu J, Cao X. Cellular and molecular regulation of innate inflammatory responses. Cell Mol Immunol (2016) 13(6):711–21. 10.1038/cmi.2016.58
    1. Park SJ, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa S, et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J Immunol (2004) 173(6):3844–54. 10.4049/jimmunol.173.6.3844
    1. Hwang W, Jung K, Jeon Y, Yun S, Kim TW, Choi I. Knockdown of the interleukin-6 receptor alpha chain of dendritic cell vaccines enhances the therapeutic potential against IL-6 producing tumors. Vaccine (2010) 29(1):34–44. 10.1016/j.vaccine.2010.10.027
    1. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med (2009) 15(1):42–9. 10.1038/nm.1905
    1. Chiossone L, Conte R, Spaggiari GM, Serra M, Romei C, Bellora F, et al. Mesenchymal Stromal Cells Induce Peculiar Alternatively Activated Macrophages Capable of Dampening Both Innate and Adaptive Immune Responses. Stem Cells (2016) 34(7):1909–21. 10.1002/stem.2369
    1. Saldana L, Bensiamar F, Valles G, Mancebo FJ, Garcia-Rey E, Vilaboa N. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther (2019) 10(1):58. 10.1186/s13287-019-1156-6
    1. Matsushita K, Takeuchi O, Standley DM, Kumagai Y, Kawagoe T, Miyake T, et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature (2009) 458(7242):1185–90. 10.1038/nature07924
    1. Suzuki HI, Arase M, Matsuyama H, Choi YL, Ueno T, Mano H, et al. MCPIP1 ribonuclease antagonizes dicer and terminates microRNA biogenesis through precursor microRNA degradation. Mol Cell (2011) 44(3):424–36. 10.1016/j.molcel.2011.09.012

Source: PubMed

3
Subscribe