Myeloid-derived suppressor cells inhibit responses of T follicular helper cells during experimental Plasmodium yoelii infection.

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • Additional Information
    • Source:
      Publisher: Federation of American Societies for Experimental Biology Country of Publication: United States NLM ID: 8804484 Publication Model: Print Cited Medium: Internet ISSN: 1530-6860 (Electronic) Linking ISSN: 08926638 NLM ISO Abbreviation: FASEB J Subsets: MEDLINE
    • Publication Information:
      Publication: 2020- : [Bethesda, Md.] : Hoboken, NJ : Federation of American Societies for Experimental Biology ; Wiley
      Original Publication: [Bethesda, Md.] : The Federation, [c1987-
    • Subject Terms:
    • Abstract:
      Malaria remains a significant global public health problem. T follicular helper (Tfh) cells, a subset of CD4 + T cells, have the capacity to regulate B cells, plasma cells, and antibody production, among other functions. Myeloid-derived suppressor cells (MDSCs) possess strong immunosuppressive abilities and can negatively regulate various immune responses. However, the role of MDSCs in inhibiting Tfh-cell responses during Plasmodium infection remains unclear. In this study, we investigated the regulatory effect of MDSCs on Tfh cell-mediated immune responses upon Plasmodium infection. We found that the numbers of MDSCs increased upon Plasmodium infection. Further mechanism study revealed that MDSC-derived Arg-1 and PD-L1 prevented Tfh cell proliferation and activation. Conversely, the addition of nor-NOHA or anti-PD-L1 monoclonal antibodies enhanced the proliferation and activation of Tfh cells, indicating that the inhibitory effect of MDSCs on Tfh cells was dependent on Arg-1 and PD-1/PD-L1. In vivo depletion of MDSCs enhanced Tfh-cell responses and antibody production, as well as relieved symptoms of infected mice and improved their survival rates. These findings provide insights into the immunosuppressive role of MDSCs in inhibiting Tfh cell immune responses and further impairing humoral immunity. Our study provides new strategies for malaria prevention and control.
      (© 2024 Federation of American Societies for Experimental Biology.)
    • References:
      WORLD HEALTH ORGANIZATION. World malaria report 2021 [M]. World Health Organization, 2021.
      Menard D, Dondorp A. Antimalarial drug resistance: a threat to malaria elimination. Cold Spring Harb Perspect Med. 2017;7(7):a025619.
      Padma TV. Combating drug resistance in malaria. Nature (London). 2023;618(7967):S26‐S28.
      Schofield L, Grau GE. Immunological processes in malaria pathogenesis. Nat Rev Immunol. 2005;5(9):722‐735.
      Su X, Lane KD, Xia L, Sá JM, Wellems TE. Plasmodium genomics and genetics: new insights into malaria pathogenesis, drug resistance, epidemiology, and evolution. Clin Microbiol Rev. 2019;32(4):e00019‐19.
      Kurup SP, Butler NS, Harty JT. T cell‐mediated immunity to malaria. Nat Rev Immunol. 2019;19(7):457‐471.
      Soon M, Haque A. Recent insights into CD4(+) Th cell differentiation in malaria. J Immunol. 2018;200(6):1965‐1975.
      Bueno LL, Morais CG, Lacerda MV, Fujiwara RT, Braga ÉM. Interleukin‐17 producing T helper cells are increased during natural Plasmodium vivax infection. Acta Trop. 2012;123(1):53‐57.
      Hansen DS, Obeng‐Adjei N, Ly A, Ioannidis LJ, Crompton PD. Emerging concepts in T follicular helper cell responses to malaria. Int J Parasitol. 2017;47(2–3):105‐110.
      Perez‐Mazliah D, Langhorne J. CD4 T‐cell subsets in malaria: TH1/TH2 revisited. Front Immunol. 2014;5:671.
      Osii RS, Otto TD, Garside P, Ndungu FM, Brewer JM. The impact of malaria parasites on dendritic cell–T cell interaction. Front Immunol. 2020;11:1597.
      Pérez‐Mazliah D, Ng DHL, Freitas do Rosário AP, et al. Disruption of IL‐21 signaling affects T cell‐B cell interactions and abrogates protective humoral immunity to malaria. PLoS Pathog. 2015;11(3):e1004715.
      Chiu CYH, Hodder AN, Lin CS, et al. Antibodies to the Plasmodium falciparum proteins MSPDBL1 and MSPDBL2 opsonize merozoites, inhibit parasite growth, and predict protection from clinical malaria. J Infect Dis. 2015;212(3):406‐415.
      Beeson JG, Chan J, Fowkes FJ. PfEMP1 as a target of human immunity and a vaccine candidate against malaria. Expert Rev Vaccines. 2013;12(2):105‐108.
      Hegde S, Leader AM, Merad M. MDSC: markers, development, states, and unaddressed complexity. Immunity (Cambridge, MA). 2021;54(5):875‐884.
      Veglia F, Perego M, Gabrilovich D. Myeloid‐derived suppressor cells coming of age. Nat Immunol. 2018;19(2):108‐119.
      Li K, Shi H, Zhang B, et al. Myeloid‐derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6(1):362.
      Grover A, Sanseviero E, Timosenko E, Gabrilovich DI. Myeloid‐derived suppressor cells: a propitious road to clinic. Cancer Discov. 2021;11(11):2693‐2706.
      Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid‐derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37(3):208‐220.
      Weber R, Groth C, Lasser S, et al. IL‐6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021;359:104254.
      Ost M, Singh A, Peschel A, Mehling R, Rieber N, Hartl D. Myeloid‐derived suppressor cells in bacterial infections. Front Cell Infect Microbiol. 2016;6:37.
      Heim CE, Vidlak D, Scherr TD, et al. Myeloid‐derived suppressor cells contribute to Staphylococcus aureus orthopedic biofilm infection. J Immunol. 2014;192(8):3778‐3792.
      Tebartz C, Horst SA, Sparwasser T, et al. A major role for myeloid‐derived suppressor cells and a minor role for regulatory T cells in immunosuppression during Staphylococcus aureus infection. J Immunol. 2015;194(3):1100‐1111.
      Yang Q, Qiu H, Xie H, et al. A Schistosoma japonicum infection promotes the expansion of myeloid‐derived suppressor cells by activating the JAK/STAT3 pathway. J Immunol. 2017;198(12):4716‐4727.
      Mukherjee S, Ghosh S, Sengupta A, et al. IL‐6 dependent expansion of inflammatory MDSCs (CD11b+ Gr‐1+) promote Th‐17 mediated immune response during experimental cerebral malaria. Cytokine. 2022;155:155910.
      Lamsfus Calle C, Fendel R, Singh A, et al. Expansion of functional myeloid‐derived suppressor cells in controlled human malaria infection. Front Immunol. 2021;12:625712.
      Zhu Y, Zhou L, Mo L, et al. Plasmodium yoelii infection enhances the expansion of myeloid‐derived suppressor cells via JAK/STAT3 pathway. J Immunol. 2024;213(2):170‐186.
      Vollbrecht T, Stirner R, Tufman A, et al. Chronic progressive HIV‐1 infection is associated with elevated levels of myeloid‐derived suppressor cells. AIDS. 2012;26(12):F31‐F37.
      Dorhoi A, Kotzé LA, Berzofsky JA, et al. Therapies for tuberculosis and AIDS: myeloid‐derived suppressor cells in focus. J Clin Invest. 2020;130(6):2789‐2799.
      Reizine F, Lesouhaitier M, Gregoire M, et al. SARS‐CoV‐2‐induced ARDS associates with MDSC expansion, lymphocyte dysfunction, and arginine shortage. J Clin Immunol. 2021;41(3):515‐525.
      Tan Y, Tang F. SARS‐CoV‐2‐mediated immune system activation and potential application in immunotherapy. Med Res Rev. 2021;41(2):1167‐1194.
      Ohl K, Tenbrock K. Reactive oxygen species as regulators of MDSC‐mediated immune suppression. Front Immunol. 2018;9:2499.
      Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN‐MDSC: their biological role and interaction with stromal cells. Semin Immunol. 2018;35:19‐28.
      Zhang Y, Wu Y, Liu H, et al. Granulocytic myeloid‐derived suppressor cells inhibit T follicular helper cells during experimental Schistosoma japonicum infection. Parasit Vectors. 2021;14(1):497.
      Wang C, Zhang N, Qi L, et al. Myeloid‐derived suppressor cells inhibit T follicular helper cell immune response in Japanese encephalitis virus infection. J Immunol. 2017;199(9):3094‐3105.
      Vijay R, Guthmiller JJ, Sturtz AJ, et al. Infection‐induced plasmablasts are a nutrient sink that impairs humoral immunity to malaria. Nat Immunol. 2020;21(7):790‐801.
      Dutta S, Tewari A, Balaji C, et al. Strain‐transcending neutralization of malaria parasite by antibodies against Plasmodium falciparum enolase. Malar J. 2018;17(1):304.
      Condamine T, Kumar V, Ramachandran IR, et al. ER stress regulates myeloid‐derived suppressor cell fate through TRAIL‐R‐mediated apoptosis. J Clin Invest. 2014;124(6):2626‐2639.
      James BR, Anderson KG, Brincks EL, et al. CpG‐mediated modulation of MDSC contributes to the efficacy of Ad5‐TRAIL therapy against renal cell carcinoma. Cancer Immunol Immunother. 2014;63(11):1213‐1227.
      Dominguez GA, Condamine T, Mony S, et al. Selective targeting of myeloid‐derived suppressor cells in cancer patients using DS‐8273a, an agonistic TRAIL‐R2 antibody. Clin Cancer Res. 2017;23(12):2942‐2950.
      de Gruijter NM, Jebson B, Rosser EC. Cytokine production by human B cells: role in health and autoimmune disease. Clin Exp Immunol. 2022;210(3):253‐262.
      Bao Y, Cao X. The immune potential and immunopathology of cytokine‐producing B cell subsets: a comprehensive review. J Autoimmun. 2014;55:10‐23.
      Vincent J, Mignot G, Chalmin F, et al. 5‐fluorouracil selectively kills tumor‐associated myeloid‐derived suppressor cells resulting in enhanced T cell–dependent antitumor immunity. Cancer Res. 2010;70(8):3052‐3061.
      Veglia F, Sanseviero E, Gabrilovich DI. Myeloid‐derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485‐498.
      Mukherjee S, Ghosh S, Bawali S, et al. Administration of soluble gp130Fc disrupts M‐1 macrophage polarization, dendritic cell activation, MDSC expansion and Th‐17 induction during experimental cerebral malaria. Int Immunopharmacol. 2023;123:110671.
      Gbedande K, Carpio VH, Stephens R. Using two phases of the CD4 T cell response to blood‐stage murine malaria to understand regulation of systemic immunity and placental pathology in Plasmodium falciparum infection. Immunol Rev. 2020;293(1):88‐114.
      White MT, Bejon P, Olotu A, et al. The relationship between RTS,S vaccine‐induced antibodies, CD4(+) T cell responses and protection against Plasmodium falciparum infection. PLoS One. 2013;8(4):e61395.
      Gray JI, Westerhof LM, MacLeod MKL. The roles of resident, central and effector memory CD4 T‐cells in protective immunity following infection or vaccination. Immunology. 2018;154(4):574‐581.
      Lumsden JM, Schwenk RJ, Rein LE, et al. Protective immunity induced with the RTS,S/AS vaccine is associated with IL‐2 and TNF‐α producing effector and central memory CD4 T cells. PLoS One. 2011;6(7):e20775.
      Pérez‐Mazliah D, Nguyen MP, Hosking C, et al. Follicular helper T cells are essential for the elimination of Plasmodium infection. EBioMedicine. 2017;24:216‐230.
      Figueiredo MM et al. T follicular helper cells regulate the activation of B lymphocytes and antibody production during Plasmodium vivax infection. PLoS Pathog. 2017;13(7):e1006484.
      Condamine T, Mastio J, Gabrilovich DI. Transcriptional regulation of myeloid‐derived suppressor cells. J Leukoc Biol. 2015;98(6):913‐922.
      Waight JD, Netherby C, Hensen ML, et al. Myeloid‐derived suppressor cell development is regulated by a STAT/IRF‐8 axis. J Clin Invest. 2013;123(10):4464‐4478.
      Pham TT et al. Hemozoin in malarial complications: more questions than answers. Trends Parasitol. 2021;37(3):226‐239.
      Kalantari P, DeOliveira RB, Chan J, et al. Dual engagement of the NLRP3 and AIM2 inflammasomes by plasmodium‐derived hemozoin and DNA during malaria. Cell Rep. 2014;6(1):196‐210.
      Xie M, Lin Z, Ji X, et al. FGF19/FGFR4‐mediated elevation of ETV4 facilitates hepatocellular carcinoma metastasis by upregulating PD‐L1 and CCL2. J Hepatol. 2023;79(1):109‐125.
      He Y, Li X, Perego M, et al. Transitory presence of myeloid‐derived suppressor cells in neonates is critical for control of inflammation. Nat Med. 2018;24(2):224‐231.
      Sercundes MK, Ortolan LS, Debone D, et al. Targeting neutrophils to prevent malaria‐associated acute lung injury/acute respiratory distress syndrome in mice. PLoS Pathog. 2016;12(12):e1006054.
      Schumak B, Klocke K, Kuepper JM, et al. Specific depletion of Ly6Chi inflammatory monocytes prevents immunopathology in experimental cerebral malaria. PLoS One. 2015;10(4):e0124080.
      Su Z, Stevenson MM. IL‐12 is required for antibody‐mediated protective immunity against blood‐stage Plasmodium chabaudi AS malaria infection in mice. J Immunol. 2002;168(3):1348‐1355.
      D Ombrain MC et al. Association of early interferon‐γ production with immunity to clinical malaria: a longitudinal study among Papua New Guinean children. Clin Infect Dis. 2008;47(11):1380‐1387.
      King T, Lamb T. Interferon‐γ: the Jekyll and Hyde of malaria. PLoS Pathog. 2015;11(10):e1005118.
      Ortega MA, Boaru DL, De Leon‐Oliva D, et al. PD‐1/PD‐L1 axis: implications in immune regulation, cancer progression, and translational applications. J Mol Med (Berl). 2024;102(8):987‐1000.
      Yamaguchi H, Hsu JM, Sun L, Wang SC, Hung MC. Advances and prospects of biomarkers for immune checkpoint inhibitors. Cell Rep Med. 2024;5(7):101621.
      Javed SA, Najmi A, Ahsan W, Zoghebi K. Targeting PD‐1/PD‐L‐1 immune checkpoint inhibition for cancer immunotherapy: success and challenges. Front Immunol. 2024;15:1383456.
      Wikenheiser DJ et al. The costimulatory molecule ICOS regulates host Th1 and follicular Th cell differentiation in response to Plasmodium chabaudi AS infection. J Immunol. 2016;196(2):778‐791.
      Faleiro R, Karunarathne DS, Horne‐Debets JM, Wykes M. The contribution of Co‐signaling pathways to anti‐malarial T cell immunity. Front Immunol. 2018;9:2926.
      Mintz MA, Cyster JG. T follicular helper cells in germinal center B cell selection and lymphomagenesis. Immunol Rev. 2020;296(1):48‐61.
      Vinuesa CG, Linterman MA, Yu D, IC ML. Follicular helper T cells. Annu Rev Immunol. 2016;34:335‐368.
      Jayakumar A, Bothwell ALM. Functional diversity of myeloid‐derived suppressor cells: the multitasking hydra of cancer. J Immunol. 2019;203(5):1095‐1103.
      Özkan B, Lim H, Park SG. Immunomodulatory function of myeloid‐derived suppressor cells during B cell‐mediated immune responses. Int J Mol Sci. 2018;19(5):1468.
      Yadav K, Shivahare R, Shaham SH, Joshi P, Sharma A, Tripathi R. Repurposing of existing therapeutics to combat drug‐resistant malaria. Biomed Pharmacother. 2021;136:111275.
      Gielecińska A, Kciuk M, Yahya EB, Ainane T, Mujwar S, Kontek R. Apoptosis, necroptosis, and pyroptosis as alternative cell death pathways induced by chemotherapeutic agents? Biochim Biophys Acta Rev Cancer. 2023;1878(6):189024.
      Kim YJ, Choi J, Choi YS. Transcriptional regulation of Tfh dynamics and the formation of immunological synapses. Exp Mol Med. 2024;56(6):1365‐1372.
      Choi J, Crotty S. Bcl6‐mediated transcriptional regulation of follicular helper T cells (TFH). Trends Immunol. 2021;42(4):336‐349.
      Yu D, Walker LSK, Liu Z, Linterman MA, Li Z. Targeting TFH cells in human diseases and vaccination: rationale and practice. Nat Immunol. 2022;23(8):1157‐1168.
    • Grant Information:
      2022YFC2304205 MOST | National Key Research and Development Program of China (NKPs); 82471791 MOST | National Natural Science Foundation of China (NSFC); 82304574 MOST | National Natural Science Foundation of China (NSFC); A2024163 Medical scientific research foundation of Guangdong province; 202301-404 The open research funds from the Sixth Affiliated Hospital of Guangzhou Medical University; Qingyuan People's Hospital; PX-66221466 Students' Innovation Ability Enhancement Plan project of Guangzhou Medical University; JCXKJS2021C08 Discipline Construction Fund of Guangzhou Medical University; GZNL2023A01009 major projects of guangzhou national laboratory; GZNL2024A01017 major projects of guangzhou national laboratory; 2024312082 Tertiary education scientific research project of Guangzhou Municipal Education Bureau
    • Contributed Indexing:
      Keywords: Arg‐1; MDSCs; PD‐L1; Tfh cells; malaria
    • Accession Number:
      0 (B7-H1 Antigen)
      0 (Programmed Cell Death 1 Receptor)
      EC 3.5.3.1 (Arginase)
      0 (Cd274 protein, mouse)
      EC 3.5.3.1 (Arg1 protein, mouse)
      0 (Pdcd1 protein, mouse)
    • Publication Date:
      Date Created: 20241204 Date Completed: 20241204 Latest Revision: 20241204
    • Publication Date:
      20241204
    • Accession Number:
      10.1096/fj.202401237R
    • Accession Number:
      39629706