Bronchus-associated lymphoid tissue-resident Foxp3+ T lymphocytes prevent antibody-mediated lung rejection

Wenjun Li, Jason M Gauthier, Ryuji Higashikubo, Hsi-Min Hsiao, Satona Tanaka, Linh Vuong, Jon H Ritter, Alice Y Tong, Brian W Wong, Ramsey R Hachem, Varun Puri, Ankit Bharat, Alexander S Krupnick, Chyi S Hsieh, William M Baldwin 3rd, Francine L Kelly, Scott M Palmer, Andrew E Gelman, Daniel Kreisel, Wenjun Li, Jason M Gauthier, Ryuji Higashikubo, Hsi-Min Hsiao, Satona Tanaka, Linh Vuong, Jon H Ritter, Alice Y Tong, Brian W Wong, Ramsey R Hachem, Varun Puri, Ankit Bharat, Alexander S Krupnick, Chyi S Hsieh, William M Baldwin 3rd, Francine L Kelly, Scott M Palmer, Andrew E Gelman, Daniel Kreisel

Abstract

Antibody-mediated rejection (AMR) is a principal cause of acute and chronic failure of lung allografts. However, mechanisms mediating this oftentimes fatal complication are poorly understood. Here, we show that Foxp3+ T cells formed aggregates in rejection-free human lung grafts and accumulated within induced bronchus-associated lymphoid tissue (BALT) of tolerant mouse lungs. Using a retransplantation model, we show that selective depletion of graft-resident Foxp3+ T lymphocytes resulted in the generation of donor-specific antibodies (DSA) and AMR, which was associated with complement deposition and destruction of airway epithelium. AMR was dependent on graft infiltration by B and T cells. Depletion of graft-resident Foxp3+ T lymphocytes resulted in prolonged interactions between B and CD4+ T cells within transplanted lungs, which was dependent on CXCR5-CXCL13. Blockade of CXCL13 as well as inhibition of the CD40 ligand and the ICOS ligand suppressed DSA production and prevented AMR. Thus, we have shown that regulatory Foxp3+ T cells residing within BALT of tolerant pulmonary allografts function to suppress B cell activation, a finding that challenges the prevailing view that regulation of humoral responses occurs peripherally. As pulmonary AMR is largely refractory to current immunosuppression, our findings provide a platform for developing therapies that target local immune responses.

Keywords: Immunology; Organ transplantation; T cells; Tolerance; Transplantation.

Conflict of interest statement

Conflict of interest: DK has a pending patent entitled “Compositions and methods for detecting CCR2 receptors” (application number 15/611,577). DK also serves on the Scientific Advisory Board of Compass Therapeutics and has received research support from Compass Therapeutics.

Figures

Figure 1. Long-term acceptance after lung transplantation…
Figure 1. Long-term acceptance after lung transplantation is associated with induction of Foxp3+ cell–rich BALT.
(A) Gross and (B) histological appearance (H&E) of BALB/c lung graft (Tx) at least 30 days after transplantation into an immunosuppressed B6 host. Arrow depicts induced BALT (n = 8). Scale bar: 100 μm. (C) PNAd staining (brown), (D) MT staining (blue), and (E) immunofluorescent staining of CCSP (red) and AcT (green) in BALB/c lung graft at least 30 days after transplantation into an immunosuppressed B6 host. Scale bars: 100 μm. (F) Intravital 2-photon (2P) imaging depicting aggregates of Foxp3+ cells in BALB/c lung graft at least 30 days after transplantation into an immunosuppressed B6 Foxp3-IRES GFP recipient (Foxp3+ cells, green; quantum dot–labeled vessels, red) (n = 3). Scale bar: 30 μm.
Figure 2. Depletion of graft-resident Foxp3 +…
Figure 2. Depletion of graft-resident Foxp3+ T cells triggers AMR.
(A) Schematic depicting experimental model. CSB, costimulation blockade. Plots and quantification of CD4+Foxp3+ cells from (B) primary (CD45.2) or (C) secondary recipient (CD45.1) in BALB/c lungs, transplanted into immunosuppressed WT B6 (CD45.2) (circles) or Foxp3-DTR B6 (CD45.2) (inverted triangles) mice and retransplanted into DT-treated B6 (CD45.1) hosts at least 30 days later. Plots are gated on live CD45.2+CD45.1–CD90.2+ and live CD45.2–CD45.1+CD90.2+ cells. (D) Plots and quantification of distribution of CD45.1 vs. CD45.2 on live CD90.2+CD4+CD8–Foxp3+ cells without and with depletion of graft-resident (CD45.2) Foxp3+ cells. Gross and histological appearance (H&E) of BALB/c lungs, transplanted into immunosuppressed WT (E and G) or Foxp3-DTR (F and H) B6 CD45.2+ mice and, at least 30 days later, retransplanted into DT-treated B6 CD45.1+ hosts (n = 4 mice per group). (I) Airway epithelium (arrow) in human lung diagnosed with AMR (H&E) (n = 11). Staining of (J and K) MT (blue), (L and M) CCSP (red), AcT (green), and (N and O) C4d (brown) in BALB/c lungs, transplanted into immunosuppressed (J, L, and N) WT or (K, M, and O) Foxp3-DTR B6 CD45.2+ mice and, at least 30 days later, retransplanted into DT-treated B6 CD45.1+ hosts. Scale bars: 100 μm. Arrows in G, J, and L point to BALT. (P) Donor-specific IgM antibody titers 7 days after retransplantation of BALB/c lungs into DT-treated B6 CD45.1+ hosts at least 30 days after initial engraftment into immunosuppressed B6 WT (red) or B6 Foxp3-DTR (blue) mice. (Q) Reactivity of serum IgM antibodies, following depletion of graft-resident Foxp3 cells, against donor (BALB/c), recipient (B6), and third-party (CBA) antigen (n = 4 mice per group). Data are expressed as mean ± SEM. Mann-Whitney U test was used to compare the means.
Figure 3. Graft-resident Foxp3 + T cells…
Figure 3. Graft-resident Foxp3+ T cells maintain lung transplant tolerance.
(A and E) Gross and (B and F) histological appearance (H&E), (C and G) MT staining (blue), and (D and H) immunofluorescent staining of CCSP (red) and AcT (green) in BALB/c lungs, initially transplanted into immunosuppressed (AD) B6 (CD45.2+) or (EH) B6 Foxp3-DTR (CD45.2+) mice and retransplanted into DT-treated secondary B6 (CD45.1+) hosts at least 30 days after primary transplantation. Secondary hosts were examined 30 days after retransplantation. Scale bars: 100 μm. Arrows in B and C point to BALT. Serum titers of donor-specific (I) IgM and (J) IgG antibodies 30 days after retransplantation of BALB/c lungs, initially transplanted into immunosuppressed B6 WT (red) or B6 Foxp3-DTR (blue) mice and, at least 30 days later, retransplanted into B6 45.1+ hosts. (K) Reactivity of serum IgG antibodies, following depletion of graft-resident Foxp3 cells, against donor (BALB/c), recipient (B6), and third-party (CBA) antigen. Data are expressed as mean ± SEM (n = 4 mice per group). Mann-Whitney U test was used to compare the means.
Figure 4. Graft-infiltrating B cells trigger AMR…
Figure 4. Graft-infiltrating B cells trigger AMR after depletion of graft-resident Foxp3+ T cells.
Activated B cells from (A and B) primary recipient (CD45.2) or (C and D) secondary recipient (CD45.1) in BALB/c lungs, transplanted into immunosuppressed (A and C) B6 (CD45.2+) (no Foxp3 depletion) or (B and D) B6 Foxp3-DTR (CD45.2+) mice (Foxp3 depletion) and, at least 30 days later, retransplanted into DT-treated B6 (CD45.1+) hosts. Plots are gated on live CD45.2+CD45.1–B220+ (donor) and live CD45.2–CD45.1+B220+ cells (recipient). Quantification of activated (E) CD45.2 and (F) CD45.1 B cells in (circles) control and (inverted triangles) Foxp3+ T cell–depleted lungs 7 days after retransplantation. (G) Gross, (H) histological appearance (H&E), (I) MT staining (blue), and (J) CCSP (red) and AcT (green) staining in BALB/c lungs, transplanted into immunosuppressed B6 Foxp3-DTR mice and, at least 30 days later, retransplanted into DT-treated B6 muMt– hosts. (K) Donor-specific IgM titers 7 days after retransplantation of BALB/c lungs into DT-treated WT (blue) or muMt– (red) B6 hosts at least 30 days after engraftment into immunosuppressed B6 Foxp3-DTR mice. (L) Gross and (M) histological appearance (H&E) of BALB/c lungs, transplanted into immunosuppressed B6 Foxp3-DTR mice and, at least 30 days later, retransplanted into DT-treated B6 AID/μS knockout hosts. Scale bars: 100 μm. (N) Donor-specific IgM titers 7 days after retransplantation of BALB/c lungs into DT-treated WT (blue) or AID/μS knockout (red) B6 hosts at least 30 days after engraftment into immunosuppressed B6 Foxp3-DTR mice. Data are expressed as mean ± SEM (n = 4 mice per group). Mann-Whitney U test was used to compare the means.
Figure 5. AMR after depletion of graft-resident…
Figure 5. AMR after depletion of graft-resident Foxp3+ T cells is dependent on graft infiltration by T cells.
Plots and histograms depicting markers characteristic of Tfh cells on CD4+ T cells derived from (A and C) primary (CD45.2) or (B and D) secondary (CD45.1) recipients in BALB/c lungs, initially transplanted into immunosuppressed (A and B) B6 (CD45.2+) or (C and D) B6 Foxp3-DTR (CD45.2+) mice and, at least 30 days later, retransplanted into DT-treated B6 (CD45.1+) hosts. Plots are gated on live CD45.2+CD45.1–CD90.2+CD4+CD8–Foxp3– and CD45.2+CD45.1+CD90.2+CD4+CD8–Foxp3– cells. Histograms are gated on CD4+ T cells that are PD-1hiCXCR5+ (bcl-6, red; isotype control, blue) (n = 4 each). Quantification of (E) CD45.2+ and (F) CD45.1+ CD4+ T cells that are PD-1hiCXCR5+ in (circles) control and (inverted triangles) Foxp3+ T cell–depleted lungs 7 days after retransplantation. (G) Gross and (H) histological appearance (H&E) and staining for (I) MT, (J) CCSP (red), and AcT (green) in BALB/c lungs, transplanted into immunosuppressed B6 Foxp3-DTR mice and, at least 30 days later, retransplanted into DT-treated B6 nude hosts. Scale bars: 100 μm. (K) Donor-specific IgM titers 7 days after retransplantation of BALB/c lungs into DT-treated WT (blue) or nude (red) B6 hosts at least 30 days after initial engraftment into immunosuppressed B6 Foxp3-DTR mice. Data are expressed as mean ± SEM (n = 4 mice per group). Mann-Whitney U test was used to compare the means.
Figure 6. Foxp3 + T lymphocyte depletion–triggered…
Figure 6. Foxp3+ T lymphocyte depletion–triggered AMR is dependent on CXCL13-mediated chemokinesis.
(A) Foxp3+ (green), B cells (blue), and CD4+ T cells (red) in BALB/c lungs at least 30 days after transplantation into immunosuppressed B6 Foxp3-IRES GFP recipient (n = 3). Scale bar: 10 μm. CXCR5+ B cells from secondary host (recipient) in BALB/c lungs, initially transplanted into immunosuppressed (B) WT or (C) Foxp3-DTR B6 (CD45.2+) recipient and, at least 30 days later, retransplanted into DT-treated B6 CD45.1+ hosts. Plots are gated on live CD45.2–CD45.1+ cells. (D) CD45.1+CXCR5+ B cells in (circles) control and (inverted triangles) Foxp3+ T cell–depleted lungs 7 days after retransplantation (n = 4 each). CXCL13 (brown) in (E) control and (F) Foxp3+ T cell–depleted grafts 7 days after retransplantation. Scale bars: 100 μm. CD4+ T cells (green) and B cells (blue) in BALB/c lungs, initially transplanted into immunosuppressed B6 Foxp3-DTR recipients and, at least 30 days later, retransplanted into B6 hosts, treated with (G) DT/control-Ig (arrows: CD4+ T–B cell interactions) or (H) DT/anti-CXCL13 (n = 2 each) (red, quantum dots). Scale bars: 20 μm. (I) Contact duration between CD4+ T and B cells, (J) CD4+ T, and (K) B cell mean square displacements and (L) CD4+ T and (M) B cell velocities within retransplanted Foxp3+ T cell–depleted BALB/c lungs with and without CXCL13 inhibition. (N) Gross, (O) histological appearance (H&E), staining for (P) MT (blue), (Q) CCSP (red), and AcT (green) in BALB/c lungs, transplanted into immunosuppressed B6 Foxp3-DTR mice and, at least 30 days later, retransplanted into DT- and anti-CXCL13–treated B6 hosts. Scale bars: 100 μm. (R) Donor-specific IgM titers after retransplantation of BALB/c lungs into DT-treated control (blue) or DT/anti-CXCL13 antibody–treated (red) B6 recipients after initial engraftment into immunosuppressed B6 Foxp3-DTR mice (n = 4 mice per group). Data are expressed as mean ± SEM. Mann-Whitney U test was used to compare the means.
Figure 7. AMR of Foxp3 + T…
Figure 7. AMR of Foxp3+ T cell–depleted lung grafts can be prevented by blocking ICOS/ICOS ligand and CD40/CD40 ligand pathways.
(A) Expression levels of ICOS, ICOS ligand (ICOSL), CD40, CD40 ligand (CD40L), and IL-21 in BALB/c lung grafts, which were initially transplanted into immunosuppressed B6 WT (CD45.2) (circles) or Foxp3-DTR B6 (CD45.2) (inverted triangles) mice and at least 30 days later retransplanted into DT-treated (days 0 and 1) B6 CD45.1 hosts. Grafts were examined 7 days after retransplantation (n = 5 each). **P < 0.01. (B) Gross appearance, (C) histological appearance (H&E), (D) MT (blue) staining, (E) and immunofluorescent staining of CCSP (red) and AcT (green) after depletion of graft-resident Foxp3+ T cells and 7 days after retransplantation into anti-ICOS ligand and anti-CD40 ligand–treated secondary hosts. Scale bars: 100 μm. (F) Serum titers of donor-specific IgM 7 days after retransplantation of BALB/c lungs, initially transplanted into immunosuppressed B6 Foxp3-DTR mice and at least 30 days later retransplanted into DT-treated WT B6 hosts that were treated with anti-ICOS ligand and anti-CD40 ligand (red) or did not receive treatment (blue) (n = 4 mice per group). Data are expressed as mean ± SEM. Mann-Whitney U test was used to compare the means.

References

    1. Chambers DC, et al. The Registry of the International Society for Heart and Lung Transplantation: Thirty-fourth Adult Lung And Heart-Lung Transplantation Report-2017; Focus Theme: Allograft ischemic time. J Heart Lung Transplant. 2017;36(10):1047–1059. doi: 10.1016/j.healun.2017.07.016.
    1. Witt CA, Puri V, Gelman AE, Krupnick AS, Kreisel D. Lung transplant immunosuppression - time for a new approach? Expert Rev Clin Immunol. 2014;10(11):1419–1421. doi: 10.1586/1744666X.2014.959499.
    1. Lakkis FG, Arakelov A, Konieczny BT, Inoue Y. Immunologic ‘ignorance’ of vascularized organ transplants in the absence of secondary lymphoid tissue. Nat Med. 2000;6(6):686–688. doi: 10.1038/76267.
    1. Zhang N, et al. Regulatory T cells sequentially migrate from inflamed tissues to draining lymph nodes to suppress the alloimmune response. Immunity. 2009;30(3):458–469. doi: 10.1016/j.immuni.2008.12.022.
    1. Krupnick AS, et al. Central memory CD8+ T lymphocytes mediate lung allograft acceptance. J Clin Invest. 2014;124(3):1130–1143. doi: 10.1172/JCI71359.
    1. Gelman AE, et al. Cutting edge: Acute lung allograft rejection is independent of secondary lymphoid organs. J Immunol. 2009;182(7):3969–3973. doi: 10.4049/jimmunol.0803514.
    1. Li W, et al. Lung transplant acceptance is facilitated by early events in the graft and is associated with lymphoid neogenesis. Mucosal Immunol. 2012;5(5):544–554. doi: 10.1038/mi.2012.30.
    1. Kelly FL, et al. Epithelial clara cell injury occurs in bronchiolitis obliterans syndrome after human lung transplantation. Am J Transplant. 2012;12(11):3076–3084. doi: 10.1111/j.1600-6143.2012.04201.x.
    1. Zeng Q, et al. B cells mediate chronic allograft rejection independently of antibody production. J Clin Invest. 2014;124(3):1052–1056. doi: 10.1172/JCI70084.
    1. Kreisel D, et al. Emergency granulopoiesis promotes neutrophil-dendritic cell encounters that prevent mouse lung allograft acceptance. Blood. 2011;118(23):6172–6182. doi: 10.1182/blood-2011-04-347823.
    1. Yamamoto S, et al. Cutting edge: Pseudomonas aeruginosa abolishes established lung transplant tolerance by stimulating B7 expression on neutrophils. J Immunol. 2012;189(9):4221–4225. doi: 10.4049/jimmunol.1201683.
    1. Onyema OO, et al. Eosinophils promote inducible NOS-mediated lung allograft acceptance. JCI Insight. 2017;2(24):e96455.
    1. Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol. 2000;164(1):183–190. doi: 10.4049/jimmunol.164.1.183.
    1. Karim M, Feng G, Wood KJ, Bushell AR. CD25+CD4+ regulatory T cells generated by exposure to a model protein antigen prevent allograft rejection: antigen-specific reactivation in vivo is critical for bystander regulation. Blood. 2005;105(12):4871–4877. doi: 10.1182/blood-2004-10-3888.
    1. Zhao Y, Li X. Cross-immune tolerance: conception and its potential significance on transplantation tolerance. Cell Mol Immunol. 2010;7(1):20–25. doi: 10.1038/cmi.2009.101.
    1. Hsiao HM, Li W, Gelman AE, Krupnick AS, Kreisel D. The role of lymphoid neogenesis in allografts. Am J Transplant. 2016;16(4):1079–1085. doi: 10.1111/ajt.13645.
    1. Thaunat O, Patey N, Morelon E, Michel JB, Nicoletti A. Lymphoid neogenesis in chronic rejection: the murderer is in the house. Curr Opin Immunol. 2006;18(5):576–579. doi: 10.1016/j.coi.2006.07.006.
    1. Baddoura FK, Nasr IW, Wrobel B, Li Q, Ruddle NH, Lakkis FG. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant. 2005;5(3):510–516.
    1. Huibers MM, et al. The composition of ectopic lymphoid structures suggests involvement of a local immune response in cardiac allograft vasculopathy. J Heart Lung Transplant. 2015;34(5):734–745. doi: 10.1016/j.healun.2014.11.022.
    1. Hiemann NE, Knosalla C, Wellnhofer E, Lehmkuhl HB, Hetzer R, Meyer R. Quilty in biopsy is associated with poor prognosis after heart transplantation. Transpl Immunol. 2008;19(3–4):209–214.
    1. Smith RN, Chang Y, Houser S, Dec GW, Grazette L. Higher frequency of high-grade rejections in cardiac allograft patients after Quilty B lesions or grade 2/4 rejections. Transplantation. 2002;73(12):1928–1932. doi: 10.1097/00007890-200206270-00014.
    1. Thaunat O, et al. Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response. Proc Natl Acad Sci U S A. 2005;102(41):14723–14728. doi: 10.1073/pnas.0507223102.
    1. Moyron-Quiroz JE, et al. Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat Med. 2004;10(9):927–934. doi: 10.1038/nm1091.
    1. Graca L, Cobbold SP, Waldmann H. Identification of regulatory T cells in tolerated allografts. J Exp Med. 2002;195(12):1641–1646. doi: 10.1084/jem.20012097.
    1. Miyajima M, et al. Early acceptance of renal allografts in mice is dependent on foxp3(+) cells. Am J Pathol. 2011;178(4):1635–1645. doi: 10.1016/j.ajpath.2010.12.024.
    1. Wan SS, Ying TD, Wyburn K, Roberts DM, Wyld M, Chadban SJ. The treatment of antibody-mediated rejection in kidney transplantation: an updated systematic review and meta-analysis. Transplantation. 2018;102(4):557–568. doi: 10.1097/TP.0000000000002049.
    1. Colvin MM, et al. Antibody-mediated rejection in cardiac transplantation: emerging knowledge in diagnosis and management: a scientific statement from the American Heart Association. Circulation. 2015;131(18):1608–1639. doi: 10.1161/CIR.0000000000000093.
    1. Witt CA, et al. Acute antibody-mediated rejection after lung transplantation. J Heart Lung Transplant. 2013;32(10):1034–1040. doi: 10.1016/j.healun.2013.07.004.
    1. Aguilar PR, et al. The role of C4d deposition in the diagnosis of antibody-mediated rejection after lung transplantation. Am J Transplant. 2018;18(4):936–944. doi: 10.1111/ajt.14534.
    1. Tikkanen JM, et al. De novo DQ donor-specific antibodies are associated with chronic lung allograft dysfunction after lung transplantation. Am J Respir Crit Care Med. 2016;194(5):596–606. doi: 10.1164/rccm.201509-1857OC.
    1. Srinivasan M, et al. Donor B-cell alloantibody deposition and germinal center formation are required for the development of murine chronic GVHD and bronchiolitis obliterans. Blood. 2012;119(6):1570–1580. doi: 10.1182/blood-2011-07-364414.
    1. Jane-Wit D, et al. Alloantibody and complement promote T cell-mediated cardiac allograft vasculopathy through noncanonical nuclear factor-κB signaling in endothelial cells. Circulation. 2013;128(23):2504–2516. doi: 10.1161/CIRCULATIONAHA.113.002972.
    1. Giangreco A, Arwert EN, Rosewell IR, Snyder J, Watt FM, Stripp BR. Stem cells are dispensable for lung homeostasis but restore airways after injury. Proc Natl Acad Sci U S A. 2009;106(23):9286–9291. doi: 10.1073/pnas.0900668106.
    1. Jaramillo A, Smith CR, Maruyama T, Zhang L, Patterson GA, Mohanakumar T. Anti-HLA class I antibody binding to airway epithelial cells induces production of fibrogenic growth factors and apoptotic cell death: a possible mechanism for bronchiolitis obliterans syndrome. Hum Immunol. 2003;64(5):521–529. doi: 10.1016/S0198-8859(03)00038-7.
    1. Fukami N, et al. Antibodies to MHC class I induce autoimmunity: role in the pathogenesis of chronic rejection. J Immunol. 2009;182(1):309–318. doi: 10.4049/jimmunol.182.1.309.
    1. Takenaka M, Tiriveedhi V, Subramanian V, Hoshinaga K, Patterson AG, Mohanakumar T. Antibodies to MHC class II molecules induce autoimmunity: critical role for macrophages in the immunopathogenesis of obliterative airway disease. PLoS ONE. 2012;7(8):e42370. doi: 10.1371/journal.pone.0042370.
    1. Khan MA, et al. CD4+ T cells and complement independently mediate graft ischemia in the rejection of mouse orthotopic tracheal transplants. Circ Res. 2011;109(11):1290–1301. doi: 10.1161/CIRCRESAHA.111.250167.
    1. Babu AN, et al. Microvascular destruction identifies murine allografts that cannot be rescued from airway fibrosis. J Clin Invest. 2007;117(12):3774–3785. doi: 10.1172/JCI32311.
    1. Luckraz H, et al. Microvascular changes in small airways predispose to obliterative bronchiolitis after lung transplantation. J Heart Lung Transplant. 2004;23(5):527–531. doi: 10.1016/j.healun.2003.07.003.
    1. Conlon TM, et al. Germinal center alloantibody responses are mediated exclusively by indirect-pathway CD4 T follicular helper cells. J Immunol. 2012;188(6):2643–2652. doi: 10.4049/jimmunol.1102830.
    1. Vu Van D, et al. Local T/B cooperation in inflamed tissues is supported by T follicular helper-like cells. Nat Commun. 2016;7:10875.
    1. Flynn R, et al. Increased T follicular helper cells and germinal center B cells are required for cGVHD and bronchiolitis obliterans. Blood. 2014;123(25):3988–3998. doi: 10.1182/blood-2014-03-562231.
    1. Leonardo SM, De Santis JL, Gehrand A, Malherbe LP, Gauld SB. Expansion of follicular helper T cells in the absence of Treg cells: implications for loss of B-cell anergy. Eur J Immunol. 2012;42(10):2597–2607. doi: 10.1002/eji.201242616.
    1. Linterman MA, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17(8):975–982. doi: 10.1038/nm.2425.
    1. Sage PT, Paterson AM, Lovitch SB, Sharpe AH. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity. 2014;41(6):1026–1039. doi: 10.1016/j.immuni.2014.12.005.
    1. Wing JB, Ise W, Kurosaki T, Sakaguchi S. Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity. 2014;41(6):1013–1025. doi: 10.1016/j.immuni.2014.12.006.
    1. Krishnamoorthy N, et al. Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma. Nat Med. 2012;18(10):1525–1530. doi: 10.1038/nm.2896.
    1. Kulkarni HS, Bemiss BC, Hachem RR. Antibody-mediated rejection in lung transplantation. Curr Transplant Rep. 2015;2(4):316–323. doi: 10.1007/s40472-015-0074-5.
    1. Okazaki M, et al. A mouse model of orthotopic vascularized aerated lung transplantation. Am J Transplant. 2007;7(6):1672–1679. doi: 10.1111/j.1600-6143.2007.01819.x.
    1. Li W, et al. Intravital 2-photon imaging of leukocyte trafficking in beating heart. J Clin Invest. 2012;122(7):2499–2508. doi: 10.1172/JCI62970.
    1. Li W, et al. Surgical technique for lung retransplantation in the mouse. J Thorac Dis. 2013;5(3):321–325.
    1. Kreisel D, et al. In vivo two-photon imaging reveals monocyte-dependent neutrophil extravasation during pulmonary inflammation. Proc Natl Acad Sci U S A. 2010;107(42):18073–18078. doi: 10.1073/pnas.1008737107.
    1. Spahn JH, et al. DAP12 expression in lung macrophages mediates ischemia/reperfusion injury by promoting neutrophil extravasation. J Immunol. 2015;194(8):4039–4048. doi: 10.4049/jimmunol.1401415.
    1. Murata K, et al. Synergistic deposition of C4d by complement-activating and non-activating antibodies in cardiac transplants. Am J Transplant. 2007;7(11):2605–2614. doi: 10.1111/j.1600-6143.2007.01971.x.

Source: PubMed

3
S'abonner