Akademska digitalna zbirka SLovenije - logo
E-viri
Celotno besedilo
Recenzirano
  • Complement activates an aut...
    Chauss, Daniel C; Freiwald, Tilo; McGregor, Reuben; Yan, Bingyu; Wang, Luopin; Nova-Lamperti, Estafania; Kumar, Dhaneshwar; Zhang, Zonghao; Teague, Heather; West, Erin; Vannella, Kevin M; Ramos-Benitez, Marcos J; Bibby, Jack; Kelly, Audrey; Malik, Amna; Freeman, Alexandra F; Schwartz, Daniella M; Portilla, Didier; Chertow, Daniel S; John, Susan; Lavender, Paul; Kemper, Claudia; Lombardi, Giovanna; Mehta, Nehal N; Cooper, Nichola; Lionakis, Michail S; Laurence, Arian; Kazemian, Majid; Afzali, Behdad

    The Journal of immunology (1950), 05/2022, Letnik: 208, Številka: 1_Supplement
    Journal Article

    Abstract Background Pro-inflammatory CD4+ T helper (Th)1 cells clear pathogens effectively but cause excessive tissue injury if not shut down appropriately. The complement (C’) system both induces Th1 differentiation and their shutdown, but the mechanisms regulating orderly shutdown remain unknown. Hypothesis C’ receptor engagement recruits transcriptional regulators essential to Th1 shutdown. Methods Multi-modal profiling of activated, or patient-derived Th cells, psoriatic skin, and SARS-CoV2-infected tissues was carried out by epigenome profiling, RNAseq, network modeling, phospho-arrays, confocal, and regulator knockdown. Results C’ receptor signaling induced the vitamin D (VitD) receptor (VDR) and CYP27B1, the enzyme that activates VitD, allowing T cells to both fully activate and respond to VitD. Active VitD shut down IFN-γ production by Th1 cells and induced IL-10. This was mediated by activation of IL-6 production by T cells and signaling through STAT3. Mechanistically, VitD reprogrammed the Th1 transcriptomes by forming super-enhancers and recruiting a transcription factor (TF) network consisting of VDR, c-JUN, STAT3, and BACH2. We mapped genome-wide targets of these TFs by CUT&RUN/Tag. As proof of principal, psoriatic skin treated with VitD induced BACH2 in Th cells, and genetic deficiency of either BACH2 or STAT3 inhibited IL-10 produced in response to VitD. Bronchoalveolar lavage fluid of COVID-19 patients, a C’-rich environment, showed excessive Th1 skewing and perturbation of the VitD-regulated program of genes. Conclusion We identified a C’-recruited autocrine VitD system as key to Th1 shutdown and indicate the potential for adjunct therapy with VitD in hyper-inflammatory syndromes, e.g. COVID-19. This work was supported by the Wellcome Trust (grant 097261/Z/11/Z to B.A.), the Crohn’s and Colitis Foundation of America (grant CCFA no. 3765 — CCFA genetics initiative to A.L.), British Heart Foundation (grant RG/13/12/30395 to G.L.), the National Institute of General Medical Sciences (R35GM138283 to M.K.), the Showalter Trust (research award to M.K.), German Research Foundation (DFG scholarship to T.F.; FR 3851/2-1), the NIDDK (DK12262401A1 to D.P.) and the National Agency of Research and Development of Chile (grant PAI79170073 to E.N.L.). Research was also supported by the National Institute for Health Research (NIHR) Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London and/or the NIHR Clinical Research Facility. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. This research was supported (in part) by the Intramural Research Programs of the NIDDK (project no. ZIA/DK075149 to B.A), the National Heart, Lung and Blood Institute (project nos. ZIA/Hl006223 to C.K. and ZIA/HL006193 to N.M.), the NIAID (project no. ZIA/AI001175 to M.S.L.) of the NIH. D.C. is supported by an NIH Office of Dietary Supplements research scholar award.