T cell-attracting CCL18 chemokine is a dominant rejection signal during limb transplantation

Thiago J Borges, Phammela Abarzua, Rodrigo B Gassen, Branislav Kollar, Mauricio Lima-Filho, Bruno T Aoyama, Diana Gluhova, Rachael A Clark, Sabina A Islam, Bohdan Pomahac, George F Murphy, Christine G Lian, Simon G Talbot, Leonardo V Riella, Thiago J Borges, Phammela Abarzua, Rodrigo B Gassen, Branislav Kollar, Mauricio Lima-Filho, Bruno T Aoyama, Diana Gluhova, Rachael A Clark, Sabina A Islam, Bohdan Pomahac, George F Murphy, Christine G Lian, Simon G Talbot, Leonardo V Riella

Abstract

Limb transplantation is a life-changing procedure for amputees. However, limb recipients have a 6-fold greater rejection rate than solid organ transplant recipients, related in part to greater immunogenicity of the skin. Here, we report a detailed immunological and molecular characterization of individuals who underwent bilateral limb transplantation at our institution. Circulating Th17 cells are increased in limb transplant recipients over time. Molecular characterization of 770 genes in skin biopsies reveals upregulation of T cell effector immune molecules and chemokines, particularly CCL18. Skin antigen-presenting cells primarily express the chemokine CCL18, which binds to the CCR8 receptor. CCL18 treatment recruits more allo-T cells to the skin xenograft in a humanized skin transplantation model, leading to signs of accelerated graft rejection. Blockade of CCR8 remarkedly decreases CCL18-induced allo-T cell infiltration. Our results suggest that targeting the CCL18:CCR8 pathway could be a promising immunosuppressive approach in transplantation.

Trial registration: ClinicalTrials.gov NCT01293214.

Keywords: CCL18; CCR8; chemokines; extremity; limb transplantation; rejection; upper extremity transplantation; vascular composite allograft.

Conflict of interest statement

The authors declare no competing interests.

© 2022 The Authors.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Analysis of circulating CD4+ and CD8+ T cell subsets from upper extremity recipients over time (A) Skin biopsies and peripheral blood were collected over time (pre-transplantation, 24 h, 1 week, and 3, 6, and 12 months after transplantation) from upper extremity transplant recipients. PBMCs were isolated for posterior flow cytometry analysis. The cartoon was created with BioRender. (B and C) Mean percentages of blood CD4+ (B) and CD8+ (C) naive cells (CCR7+CD45RA+), central memory T cells (TCM cells; CCR7+CD45RA−), effector memory T cells (TEM cells; CCR7−CD45RA−), and effector memory RA cells (TEMRA cells; CCR7−CD45RA+) before transplantation and 6 and 12 months after transplantation, represented as pie charts. Data are from all three individuals. (D) Representative contour plots of gating of T helper (Th) 1 (CD4+CXCR3+CCR6−), Th2 (CD4+CXCR3−CCR6−), and Th17 (CD4+CXCR3−CCR6+) cells. (E) Mean percentages of Th1, Th2, and Th17 cells from all three individuals over time. (F–H) Mean percentages of circulating Th17 cells before transplantation and 12 months after transplantation. Statistic by paired t test, ∗p +CD25+CD127−/low) and (H) T follicular helper (Tfh) cells (CD4+CXCR5+PD-1+) from all three individuals over time. Graphs are displayed as mean ± SD at each time point examined. See also Figure 1, Figure 2, Figure 3, Figure 4, Figure 5, Figure 6and S2.
Figure 2
Figure 2
Characterization of CD4+ and CD8+ T cell subsets from allografts and native skin of upper extremity recipients (A–F) Mean percentages per skin area of infiltrating (A) CD4+ TEM cells (CCR7−CD45RA−), (B) Th1 cells (CD4+CXCR3+CCR6−), (C) Th17 cells (CD4+CXCR3−CCR6+), (D) total CD8+ cells, (E) CD8+ TEM cells, and (F) CD8+ TEMRA cells (CCR7−CD45RA+) from the allografts and adjacent native skins. Data are from all three individuals and represented as mean ± SD; statistics by paired t test. (G) Representative H&E staining (left) and immunofluorescence of CD4+ and CD8+ cells (right) from the allograft and adjacent native skin of the same upper extremity transplant recipient; 200× (scale bars, 100 μm). See also Figure S3.
Figure 3
Figure 3
Clinical and histopathological aspects in upper extremity allograft rejection with correlation to peripheral T cell populations (A and B) Clinical photographs of an upper extremity transplant recipient and (B) corresponding H&E graft staining during clinical cellular rejection episodes with graft erythema and edema (grades 2–3) compared with mild rejection on surveillance biopsy (grade 1) without significant erythema or edema. Grade 3 rejection retains lymphocytic vasculopathy (bottom right panel) but also shows epithelial apoptosis associated with lymphoid exocytosis (circled in higher magnification, 400×). Left images, 40×; right images, 400× (scale bars, 50 μm). (C–I) Percentages and (D, F, H, and J) absolute numbers of circulating total CD8+, CD8+ TEMRA, CD4+ TEMRA, and Treg cells at rejection (grades 2–3, n = 6) and nonrejection (grade 0, n = 6) events. Percentages of (K) IFN-γ+, (L) IL-4+, (M) IL-17+ CD4+ T cells, and (N) IFN-γ+ CD8+ T cells from peripheral blood at rejection and nonrejection time points. (C–N) Data are from all three individuals and represented as mean ± SD; statistics by paired t test. Rejection time points included samples from 1 week to 3 years after transplantation and nonrejection time points from 1 month to 4 years after transplantation.
Figure 4
Figure 4
Unique gene expression signature in human upper extremity transplant rejection is associated with chemokines-related genes (A) Heatmap of the 57 differentially expressed genes (DEGs) in rejection (grades 2–3, n = 10) compared with nonrejection (grades 0–1, n = 7) skin biopsies (log2 fold change of genes assessed were transformed into Z scores). (B) Unsupervised principal-component analysis of the top 57 DEGs, clustering the samples with rejection compared with nonrejection events, except by one resolving rejection. (C) Top 8 Gene Ontology (GO) biological process terms enriched among the 57 DEGs in rejection biopsies compared with nonrejection. Statistics used Fisher’s one-tailed test with Benjamini-Hochberg false discovery rate (FDR; p value) for multiple testing correction. For all analyses, rejection biopsies included samples from 1 month to 3 years after transplantation and nonrejection biopsies from 1 month to 4.5 years after transplantation.
Figure 5
Figure 5
Chemokine-mediated signaling, T cell effector molecules, and immune checkpoints are upregulated in the limb transplant skin microenvironment during rejection (A) Volcano plot showing DEGs in rejection in relation to nonrejection. Log2 fold change is represented on the x axis, and the y axis displays −log10 of each gene’s p value. (B–E) Normalized expression of genes associated with (B) chemokines and chemokine-mediated signaling, (C) T cell co-stimulation, (D) immune checkpoints, and (E) effector molecules. Boxplots represent mean values with whiskers of maximum and minimum values. Statistics are represented by FDR p values. For all analyses, rejection biopsies (n = 10; 1 month to 3 years after transplantation) and nonrejection biopsies (n = 7; 1 month to 4.5 years after transplantation).
Figure 6
Figure 6
CCL18 attracts human allo-T cells to human skin xenografts (A and B) Discarded human skin was transplanted into NSG recipient mice. Mice were injected weekly with anti-Gr1 to reduce local inflammation and establish an intact vasculature (A). Twenty-eight days after transplantation, recipients were injected with 20 × 106 allo-PBMCs. Seven days thereafter, 300 ng of CCL18 or PBS 1× was injected subcutaneously into the skin xenografts for 10 consecutive days. Recipient animals were treated with anti-CCR8 or isotype control daily. Skin xenografts and peripheral blood were analyzed by histology and flow cytometry on day 45 after transplantation. (B–D) Representative macroscopic images of skin xenografts, H&E graft staining, and immunohistochemical staining for human CD31, human CD3, or human CD45 after the CCL18 injections into animals (A) injected (B) or not injected (C) with human PBMCs. Shown are absolute counts of human CD31+ vessels. (E and F) Human CD3+ cells (E) and human CD45+ cells (F) in the skin xenografts, 100×. The number of positive cells was quantified from two representative 400× fields from each transplanted xenograft. Statistics by one-way ANOVA with Tukey’s post-test. (G–J) Representative contour plot and absolute numbers of skin-infiltrating (G) CD4+ and CD8+ cells, (H) skin CD8+CLA+ and CD4+CLA+ cells, (I) skin CD8+CCR8+ and CD4+CCR8+ cells, and (J) Th1, Th17, and Th2 in PBS-, CCL18−, or CCL18+ anti-CCR8-treated groups that received human PBMCs. All data were normalized by square centimeter of tissue. Data represent a pool of two independent experiments (n = 4–6 animals per group) and are represented as mean ± SD. Statistics by one-way ANOVA with Tukey’s post-test. See also Figure S4.

References

    1. Murray J.E. Organ transplantation (skin, kidney, heart) and the plastic surgeon. Plast. Reconstr. Surg. 1971;47:425–431.
    1. Kollar B., Pomahac B., Riella L.V. Novel immunological and clinical insights in vascularized composite allotransplantation. Curr. Opin. Organ Transpl. 2019;24:42–48.
    1. Hein R.E., Ruch D.S., Klifto C.S., Leversedge F.J., Mithani S.K., Pidgeon T.S., Richard M.J., Cendales L.C. Hand transplantation in the United States: a review of the organ procurement and transplantation network/united network for organ sharing database. Am. J. Transpl. 2019;20:1417–1423.
    1. Islam S.A., Ling M.F., Leung J., Shreffler W.G., Luster A.D. Identification of human CCR8 as a CCL18 receptor. J. Exp. Med. 2013;210:1889–1898.
    1. Günther C., Bello-Fernandez C., Kopp T., Kund J., Carballido-Perrig N., Hinteregger S., Fassl S., Schwärzler C., Lametschwandtner G., Stingl G., et al. CCL18 is expressed in atopic dermatitis and mediates skin homing of human memory T cells. J. Immunol. 2005;174:1723–1728.
    1. Park C.O., Lee H.J., Lee J.H., Wu W.H., Chang N.S., Hua L., Lee M.G., Lee K.H. Increased expression of CC chemokine ligand 18 in extrinsic atopic dermatitis patients. Exp. Dermatol. 2008;17:24–29.
    1. Vieyra-Garcia P., Crouch J.D., O’Malley J.T., Seger E.W., Yang C.H., Teague J.E., Vromans A.M., Gehad A., Win T.S., Yu Z., et al. Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma. JCI Insight. 2019;4:e124233.
    1. Weaver C.T., Elson C.O., Fouser L.A., Kolls J.K. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu. Rev. Pathol. Mech. Dis. 2013;8:477–512.
    1. Liu T., Li S., Ying S., Tang S., Ding Y., Li Y., Qiao J., Fang H. The IL-23/IL-17 pathway in inflammatory skin diseases: from bench to bedside. Front. Immunol. 2020;11:2971.
    1. Cendales L.C., Kanitakis J., Schneeberger S., Burns C., Ruiz P., Landin L., Remmelink M., Hewitt C.W., Landgren T., Lyons B., et al. The Banff 2007 working classification of skin-containing composite tissue allograft pathology. Am. J. Transplant. 2008;8:1396–1400.
    1. Tasigiorgos S., Kollar B., Turk M., Perry B., Alhefzi M., Kiwanuka H., Nizzi M.-C., Marty F.M., Chandraker A., Tullius S.G., et al. Five-Year follow-up after face transplantation. N. Engl. J. Med. 2019;380:2579–2581.
    1. Günther C., Zimmermann N., Berndt N., Grosser M., Stein A., Koch A., Meurer M. Up-regulation of the chemokine CCL18 by macrophages is a potential immunomodulatory pathway in cutaneous T-cell lymphoma. Am. J. Pathol. 2011;179:1434–1442.
    1. He H., Suryawanshi H., Morozov P., Gay-Mimbrera J., Del Duca E., Kim H.J., Kameyama N., Estrada Y., Der E., Krueger J.G., et al. Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J. Allergy Clin. Immunol. 2020;145:1615–1628.
    1. Reynolds G., Vegh P., Fletcher J., Poyner E.F.M., Stephenson E., Goh I., Botting R.A., Huang N., Olabi B., Dubois A., et al. Developmental cell programs are co-opted in inflammatory skin disease. Science. 2021;371:eaba6500.
    1. Borges T.J., O’Malley J.T., Wo L., Murakami N., Smith B., Azzi J., Tripathi S., Lane J.D., Bueno E.M., Clark R.A., et al. Codominant role of interferon-γ– and interleukin-17–producing T cells during rejection in full facial transplant recipients. Am. J. Transpl. 2016;16:2158–2171.
    1. Krezdorn N., Lian C.G., Wells M., Wo L., Tasigiorgos S., Xu S., Borges T.J., Frierson R.M., Stanek E., Riella L.V., et al. Chronic rejection of human face allografts. Am. J. Transpl. 2019;19:1168–1177.
    1. Chung B.H., Kim K.W., Kim B.-M., Doh K.C., Cho M.-L., Yang C.W. Increase of Th17 cell phenotype in kidney transplant recipients with chronic allograft dysfunction. PLoS One. 2015;10:e0145258.
    1. Nova-Lamperti E., Romano M., Christakoudi S., Runglall M., McGregor R., Mobillo P., Kamra Y., Tsui T.-L., Norris S., John S., et al. Reduced TCR signaling contributes to impaired Th17 responses in tolerant kidney transplant recipients. Transplantation. 2018;102:e10–e17.
    1. Lian C.G., Bueno E.M., Granter S.R., Laga A.C., Saavedra A.P., Lin W.M., Susa J.S., Zhan Q., Chandraker A.K., Tullius S.G., et al. Biomarker evaluation of face transplant rejection: association of donor T cells with target cell injury. Mod. Pathol. 2014;27:788–799.
    1. Wu L., Chen X., Zhao J., Martin B., Zepp J.A., Ko J.S., Gu C., Cai G., Ouyang W., Sen G., et al. A novel IL-17 signaling pathway controlling keratinocyte proliferation and tumorigenesis via the TRAF4-ERK5 axis. J. Exp. Med. 2015;212:1571–1587.
    1. Clark R.A., Chong B., Mirchandani N., Brinster N.K., Yamanaka K.-I., Dowgiert R.K., Kupper T.S. The vast majority of CLA+ T cells are resident in normal skin. J. Immunol. 2006;176:4431–4439.
    1. Heath W.R., Carbone F.R. The skin-resident and migratory immune system in steady state and memory: innate lymphocytes, dendritic cells and T cells. Nat. Immunol. 2013;14:978–985.
    1. Etra J.W., Shores J.T., Sander I.B., Brandacher G., Lee W.P.A. Trauma-induced rejection in vascularized composite allotransplantation. Ann. Surg. 2020;271:e113–e114.
    1. Lopdrup R.G., Turk M., Win T.S., Marty F.M., Molway D., Tullius S.G., Pomahac B., Talbot S.G. Seasonal variability precipitating hand transplant rejection? Transplantation. 2017;101:e313.
    1. Tong P.L., Roediger B., Kolesnikoff N., Biro M., Tay S.S., Jain R., Shaw L.E., Grimbaldeston M.A., Weninger W. The skin immune atlas: three-dimensional analysis of cutaneous leukocyte subsets by multiphoton microscopy. J. Invest. Dermatol. 2015;135:84–93.
    1. Hautz T., Zelger B., Grahammer J., Krapf C., Amberger A., Brandacher G., Landin L., Pratschke J., Margreiter R., Schneeberger S. Molecular markers and targeted therapy of skin rejection in composite tissue allotransplantation. Am. J. Transpl. 2010;10:1200–1209.
    1. Dai H., Lan P., Zhao D., Abou-Daya K., Liu W., Chen W., Friday A.J., Williams A.L., Sun T., Chen J., et al. PIRs mediate innate myeloid cell memory to nonself MHC molecules. Science. 2020;368:1122–1127.
    1. Schaerli P., Ebert L., Willimann K., Blaser A., Roos R.S., Loetscher P., Moser B. A skin-selective homing mechanism for human immune surveillance T Cells. J. Exp. Med. 2004;199:1265–1275.
    1. Kim H.O., Cho S.I., Chung B.Y., Ahn H.K., Park C.W., Lee C.H. Expression of CCL1 and CCL18 in atopic dermatitis and psoriasis. Clin. Exp. Dermatol. 2012;37:521–526.
    1. Goebeler M., Trautmann A., Voss A., Bröcker E.B., Toksoy A., Gillitzer R. Differential and sequential expression of multiple chemokines during elicitation of allergic contact hypersensitivity. Am. J. Pathol. 2001;158:431–440.
    1. Schutyser E. Involvement of CC chemokine ligand 18 (CCL18) in normal and pathological processes. J. Leukoc. Biol. 2005;78:14–26.
    1. Colantonio L., Iellem A., Sinigaglia F., D’Ambrosio D. Skin-homing CLA+ T cells and regulatory CD25+ T cells represent major subsets of human peripheral blood memory T cells migrating in response to CCL1/I-309. Eur. J. Immunol. 2002;32:3506–3514.
    1. Kollar B., Shubin A., Borges T.J., Tasigiorgos S., Win T.S., Lian C.G., Dillon S.T., Gu X., Wyrobnik I., Murphy G.F., et al. Increased levels of circulating MMP3 correlate with severe rejection in face transplantation. Sci. Rep. 2018;8:14915.
    1. Kollar B., Uffing A., Borges T.J., Shubin A.V., Aoyama B.T., Dagot C., Haug V., Kauke M., Safi A.F., Talbot S.G., et al. MMP3 is a non-invasive biomarker of rejection in skin-bearing vascularized composite allotransplantation: a multicenter validation study. Front. Immunol. 2019;10:2771.
    1. Watarai Y., Koga S., Paolone D.R., Engeman T.M., Tannenbaum C., Hamilton T.A., Fairchild R.L. Intraallograft chemokine RNA and protein during rejection of MHC-matched/multiple minor histocompatibility-disparate skin grafts. J. Immunol. 2000;164:6027–6033.
    1. Hricik D.E., Nickerson P., Formica R.N., Poggio E.D., Rush D., Newell K.A., Goebel J., Gibson I.W., Fairchild R.L., Riggs M., et al. Multicenter validation of urinary CXCL9 as a risk-stratifying biomarker for kidney transplant injury. Am. J. Transpl. 2013;13:2634–2644.
    1. Ho J., Schaub S., Wiebe C., Gao A., Wehmeier C., Koller M.T., Hirsch H.H., Hopfer H., Nickerson P., Hirt-Minkowski P. Urinary CXCL10 chemokine is associated with alloimmune and virus compartment-specific renal allograft inflammation. Transplantation. 2018;102:521–529.
    1. Kaminski M.M., Alcantar M.A., Lape I.T., Greensmith R., Huske A.C., Valeri J.A., Marty F.M., Klämbt V., Azzi J., Akalin E., et al. A CRISPR-based assay for the detection of opportunistic infections post-transplantation and for the monitoring of transplant rejection. Nat. Biomed. Eng. 2020;4:601–609.
    1. Christofidou-Solomidou M., Longley B.J., Whitaker-Menezes D., Albelda S.M., Murphy G.F. Human skin/SCID mouse chimeras as an in vivo model for human cutaneous mast cell hyperplasia. J. Invest. Dermatol. 1997;109:102–107.
    1. Watanabe R., Gehad A., Yang C., Scott L.L., Teague J.E., Schlapbach C., Elco C.P., Huang V., Matos T.R., Kupper T.S., et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci. Transl. Med. 2015;7:279ra39.
    1. Racki W.J., Covassin L., Brehm M., Pino S., Ignotz R., Dunn R., Laning J., Graves S.K., Rossini A.A., Shultz L.D., et al. NOD-scid IL2rgamma(null) mouse model of human skin transplantation and allograft rejection. Transplantation. 2010;89:527–536.
    1. Borges T.J., Murakami N., Machado F.D., Murshid A., Lang B.J., Lopes R.L., Bellan L.M., Uehara M., Antunes K.H., Pérez-Saéz M.J., et al. March1-dependent modulation of donor MHC II on CD103+ dendritic cells mitigates alloimmunity. Nat. Commun. 2018;9:3482.
    1. Metsalu T., Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using principal component analysis and heatmap. Nucleic Acids Res. 2015;43:W566–W570.
    1. Mi H., Muruganujan A., Ebert D., Huang X., Thomas P.D. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 2019;47:D419–D426.

Source: PubMed

3
S'abonner