دورية أكاديمية

Posttransplant Tertiary Lymphoid Organs.

التفاصيل البيبلوغرافية
العنوان: Posttransplant Tertiary Lymphoid Organs.
المؤلفون: Ruddle NH; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT.
المصدر: Transplantation [Transplantation] 2024 May 01; Vol. 108 (5), pp. 1090-1099. Date of Electronic Publication: 2023 Nov 02.
نوع المنشور: Journal Article; Review; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov't
اللغة: English
بيانات الدورية: Publisher: Lippincott Williams & Wilkins Country of Publication: United States NLM ID: 0132144 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1534-6080 (Electronic) Linking ISSN: 00411337 NLM ISO Abbreviation: Transplantation Subsets: MEDLINE
أسماء مطبوعة: Publication: Hagerstown, MD : Lippincott Williams & Wilkins
Original Publication: Baltimore, Williams & Wilkins.
مواضيع طبية MeSH: Tertiary Lymphoid Structures*/immunology , Tertiary Lymphoid Structures*/pathology , Graft Rejection*/immunology , Graft Rejection*/prevention & control , Organ Transplantation*/adverse effects, Humans ; Animals
مستخلص: Tertiary lymphoid organs (TLOs), also known as tertiary or ectopic lymphoid structures or tissues, are accumulations of lymphoid cells in sites other than canonical lymphoid organs, that arise through lymphoid neogenesis during chronic inflammation in autoimmunity, microbial infection, cancer, aging, and transplantation, the focus of this review. Lymph nodes and TLOs are compared regarding their cellular composition, organization, vascular components, and migratory signal regulation. These characteristics of posttransplant TLOs (PT-TLOs) are described with individual examples in a wide range of organs including heart, kidney, trachea, lung, artery, skin, leg, hand, and face, in many species including human, mouse, rat, and monkey. The requirements for induction and maintenance of TLOs include sustained exposure to autoantigens, alloantigens, tumor antigens, ischemic reperfusion, nephrotoxic agents, and aging. Several staging schemes have been put forth regarding their function in organ rejection. PT-TLOs most often are associated with organ rejection, but in some cases contribute to tolerance. The role of PT-TLOs in cancer is considered in the case of immunosuppression. Furthermore, TLOs can be associated with development of lymphomas. Challenges for PT-TLO research are considered regarding staging, imaging, and opportunities for their therapeutic manipulation to inhibit rejection and encourage tolerance.
Competing Interests: The author declares no conflict of interest.
(Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
References: Alsughayyir J, Pettigrew GJ, Motallebzadeh R. Spoiling for a fight: B lymphocytes as initiator and effector populations within tertiary lymphoid organs in autoimmunity and transplantation. Front Immunol. 2017;8:1639.
Drayton DL, Liao S, Mounzer RH, et al. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7:344–353.
Ruddle NH. High endothelial venules and lymphatic vessels in tertiary lymphoid organs: characteristics, functions, and regulation. Front Immunol. 2016;7:491.
Sautes-Fridman C, Petitprez F, Calderaro J, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307–325.
Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375:eabf9419.
Stranford S, Ruddle NH. Follicular dendritic cells, conduits, lymphatic vessels, and high endothelial venules in tertiary lymphoid organs: parallels with lymph node stroma. Front Immunol. 2012;3:350.
Ruddle NH. Basics of inducible lymphoid organs. Curr Top Microbiol Immunol. 2020;426:1–19.
Koenig A, Thaunat O. Lymphoid neogenesis and tertiary lymphoid organs in transplanted organs. Front Immunol. 2016;7:646.
Liao S, Ruddle NH. Synchrony of high endothelial venules and lymphatic vessels revealed by immunization. J Immunol. 2006;177:3369–3379.
Brown K, Sacks SH, Wong W. Tertiary lymphoid organs in renal allografts can be associated with donor-specific tolerance rather than rejection. Eur J Immunol. 2011;41:89–96.
Thaunat O, Field AC, Dai J, et al. Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response. Proc Natl Acad Sci U S A. 2005;102:14723–14728.
Motallebzadeh R, Rehakova S, Conlon TM, et al. Blocking lymphotoxin signaling abrogates the development of ectopic lymphoid tissue within cardiac allografts and inhibits effector antibody responses. FASEB J. 2012;26:51–62.
Penaranda C, Tang Q, Ruddle NH, et al. Prevention of diabetes by FTY720-mediated stabilization of peri-islet tertiary lymphoid organs. Diabetes. 2010;59:1461–1468.
Luo R, Cheng Y, Chang D, et al. Tertiary lymphoid organs are associated with the progression of kidney damage and regulated by interleukin-17A. Theranostics. 2021;11:117–131.
Toppila S, Paavonen T, Nieminen MS, et al. Endothelial L-selectin ligands are likely to recruit lymphocytes into rejecting human heart transplants. Am J Pathol. 1999;155:1303–1310.
Sato Y, Boor P, Fukuma S, et al. Developmental stages of tertiary lymphoid tissue reflect local injury and inflammation in mouse and human kidneys. Kidney Int. 2020;98:448–463.
Turley S, Poirot L, Hattori M, et al. Physiological beta cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J Exp Med. 2003;198:1527–1537.
Connolly KA, Kuchroo M, Venkat A, et al. A reservoir of stem-like CD8(+) T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci Immunol. 2021;6:eabg7836.
Hua Y, Vella G, Rambow F, et al. Cancer immunotherapies transition endothelial cells into HEVs that generate TCF1(+) T lymphocyte niches through a feed-forward loop. Cancer Cell. 2022;40:1600–1618.e10.
Kirveskari J, Paavonen T, Hayry P, et al. De novo induction of endothelial L-selectin ligands during kidney allograft rejection. J Am Soc Nephrol. 2000;11:2358–2365.
Baddoura FK, Nasr IW, Wrobel B, et al. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant. 2005;5:510–516.
Di Carlo E, D’Antuono T, Contento S, et al. Quilty effect has the features of lymphoid neogenesis and shares CXCL13-CXCR5 pathway with recurrent acute cardiac rejections. Am J Transplant. 2007;7:201–210.
Kerjaschki D, Regele HM, Moosberger I, et al. Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates. J Am Soc Nephrol. 2004;15:603–612.
Abou-Daya KI, Tieu R, Zhao D, et al. Resident memory T cells form during persistent antigen exposure leading to allograft rejection. Sci Immunol. 2021;6:eabc8122.
Rosales IA, Yang C, Farkash EA, et al. Novel intragraft regulatory lymphoid structures in kidney allograft tolerance. Am J Transplant. 2022;22:705–716.
Sato M, Hirayama S, Hwang DM, et al. The role of intrapulmonary de novo lymphoid tissue in obliterative bronchiolitis after lung transplantation. J Immunol. 2009;182:7307–7316.
Cai J, Deng J, Gu W, et al. Impact of local alloimmunity and recipient cells in transplant arteriosclerosis. Circ Res. 2020;127:974–993.
Nasr IW, Reel M, Oberbarnscheidt MH, et al. Tertiary lymphoid tissues generate effector and memory T cells that lead to allograft rejection. Am J Transplant. 2007;7:1071–1079.
Hautz T, Zelger BG, Nasr IW, et al. Lymphoid neogenesis in skin of human hand, nonhuman primate, and rat vascularized composite allografts. Transpl Int. 2014;27:966–976.
Stuht S, Gwinner W, Franz I, et al. Lymphatic neoangiogenesis in human renal allografts: results from sequential protocol biopsies. Am J Transplant. 2007;7:377–384.
de Leur K, Clahsen-van Groningen MC, van den Bosch TPP, et al. Characterization of ectopic lymphoid structures in different types of acute renal allograft rejection. Clin Exp Immunol. 2018;192:224–232.
Zhuang Q, Liu Q, Divito SJ, et al. Graft-infiltrating host dendritic cells play a key role in organ transplant rejection. Nat Commun. 2016;7:12623.
Denton AE, Carr EJ, Magiera LP, et al. Embryonic FAP(+) lymphoid tissue organizer cells generate the reticular network of adult lymph nodes. J Exp Med. 2019;216:2242–2252.
De Togni P, Goellner J, Ruddle NH, et al. Pillars article: abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science. 1994. 264: 703-707. J Immunol. 2014;192:2010–2014.
Koni PA, Sacca R, Lawton P, et al. Distinct roles in lymphoid organogenesis for lymphotoxins alpha and beta revealed in lymphotoxin beta-deficient mice. Immunity. 1997;6:491–500.
Barone F, Gardner DH, Nayar S, et al. Stromal fibroblasts in tertiary lymphoid structures: a novel target in chronic inflammation. Front Immunol. 2016;7:477.
Nayar S, Campos J, Smith CG, et al. Immunofibroblasts are pivotal drivers of tertiary lymphoid structure formation and local pathology. Proc Natl Acad Sci U S A. 2019;116:13490–13497.
Thaunat O, Patey N, Caligiuri G, et al. Chronic rejection triggers the development of an aggressive intragraft immune response through recapitulation of lymphoid organogenesis. J Immunol. 2010;185:717–728.
Sato Y, Mii A, Hamazaki Y, et al. Heterogeneous fibroblasts underlie age-dependent tertiary lymphoid tissues in the kidney. JCI Insight. 2016;1:e87680.
Drayton DL, Ying X, Lee J, et al. Ectopic LT alpha beta directs lymphoid organ neogenesis with concomitant expression of peripheral node addressin and a HEV-restricted sulfotransferase. J Exp Med. 2003;197:1153–1163.
Kratz A, Campos-Neto A, Hanson MS, et al. Chronic inflammation caused by lymphotoxin is lymphoid neogenesis. J Exp Med. 1996;183:1461–1472.
Luther SA, Lopez T, Bai W, et al. BLC expression in pancreatic islets causes B cell recruitment and lymphotoxin-dependent lymphoid neogenesis. Immunity. 2000;12:471–481.
Meier D, Bornmann C, Chappaz S, et al. Ectopic lymphoid-organ development occurs through interleukin 7-mediated enhanced survival of lymphoid-tissue-inducer cells. Immunity. 2007;26:643–654.
Kim HJ, Kammertoens T, Janke M, et al. Establishment of early lymphoid organ infrastructure in transplanted tumors mediated by local production of lymphotoxin alpha and in the combined absence of functional B and T cells. J Immunol. 2004;172:4037–4047.
Kim MY, McConnell FM, Gaspal FM, et al. Function of CD4+CD3- cells in relation to B- and T-zone stroma in spleen. Blood. 2007;109:1602–1610.
Scandella E, Bolinger B, Lattmann E, et al. Restoration of lymphoid organ integrity through the interaction of lymphoid tissue-inducer cells with stroma of the T cell zone. Nat Immunol. 2008;9:667–675.
Ware CF, Crowe PD, Grayson MH, et al. Expression of surface lymphotoxin and tumor necrosis factor on activated T, B, and natural killer cells. J Immunol. 1992;149:3881–3888.
Moussion C, Girard JP. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature. 2011;479:542–546.
Marinkovic T, Garin A, Yokota Y, et al. Interaction of mature CD3+CD4+ T cells with dendritic cells triggers the development of tertiary lymphoid structures in the thyroid. J Clin Invest. 2006;116:2622–2632.
Grabner R, Lotzer K, Dopping S, et al. Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE-/- mice. J Exp Med. 2009;206:233–248.
Sacca R, Cuff CA, Lesslauer W, et al. Differential activities of secreted lymphotoxin-alpha3 and membrane lymphotoxin-alpha1beta2 in lymphotoxin-induced inflammation: critical role of TNF receptor 1 signaling. J Immunol. 1998;160:485–491.
Deteix C, Attuil-Audenis V, Duthey A, et al. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol. 2010;184:5344–5351.
Murakami J, Shimizu Y, Kashii Y, et al. Functional B-cell response in intrahepatic lymphoid follicles in chronic hepatitis C. Hepatology. 1999;30:143–150.
Orloff SL, Hwee YK, Kreklywich C, et al. Cytomegalovirus latency promotes cardiac lymphoid neogenesis and accelerated allograft rejection in CMV naive recipients. Am J Transplant. 2011;11:45–55.
Ichii O, Hosotani M, Masum MA, et al. Close association between altered urine-urothelium barrier and tertiary lymphoid structure formation in the renal pelvis during nephritis. J Am Soc Nephrol. 2022;33:88–107.
Adams AB, Williams MA, Jones TR, et al. Heterologous immunity provides a potent barrier to transplantation tolerance. J Clin Invest. 2003;111:1887–1895.
Boros P, Bromberg JS. New cellular and molecular immune pathways in ischemia/reperfusion injury. Am J Transplant. 2006;6:652–658.
Liao S, Bentley K, Lebrun M, et al. Transgenic LacZ under control of Hec-6st regulatory sequences recapitulates endogenous gene expression on high endothelial venules. Proc Natl Acad Sci U S A. 2007;104:4577–4582.
Singh P, Coskun ZZ, Goode C, et al. Lymphoid neogenesis and immune infiltration in aged liver. Hepatology. 2008;47:1680–1690.
Camell CD, Gunther P, Lee A, et al. Aging induces an Nlrp3 inflammasome-dependent expansion of adipose B cells that impairs metabolic homeostasis. Cell Metab. 2019;30:1024–1039.e6.
Akhavanpoor M, Gleissner CA, Akhavanpoor H, et al. Adventitial tertiary lymphoid organ classification in human atherosclerosis. Cardiovasc Pathol. 2018;32:8–14.
Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2018;7:e1378844.
Lee YH, Sato Y, Saito M, et al. Advanced tertiary lymphoid tissues in protocol biopsies are associated with progressive graft dysfunction in kidney transplant recipients. J Am Soc Nephrol. 2022;33:186–200.
Picarella DE, Kratz A, Li CB, et al. Insulitis in transgenic mice expressing tumor necrosis factor beta (lymphotoxin) in the pancreas. Proc Natl Acad Sci U S A. 1992;89:10036–10040.
Stott DI, Hiepe F, Hummel M, et al. Antigen-driven clonal proliferation of B cells within the target tissue of an autoimmune disease. The salivary glands of patients with Sjogren’s syndrome. J Clin Invest. 1998;102:938–946.
Gause A, Gundlach K, Carbon G, et al. Analysis of VH gene rearrangements from synovial B cells of patients with rheumatoid arthritis reveals infiltration of the synovial membrane by memory B cells. Rheumatol Int. 1997;17:145–150.
Cheng J, Torkamani A, Grover RK, et al. Ectopic B-cell clusters that infiltrate transplanted human kidneys are clonal. Proc Natl Acad Sci U S A. 2011;108:5560–5565.
Zhang H, Cavazzoni CB, Hanson BL, et al. Transcriptionally distinct B cells infiltrate allografts after kidney transplantation. Transplantation. 2023;107:e47–e57.
Kuerten S, Schickel A, Kerkloh C, et al. Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol. 2012;124:861–873.
McMahon EJ, Bailey SL, Castenada CV, et al. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat Med. 2005;11:335–339.
Suciu-Foca N, Harris PE, Cortesini R. Intramolecular and intermolecular spreading during the course of organ allograft rejection. Immunol Rev. 1998;164:241–246.
Moreso F, Seron D, O’Valle F, et al. Immunephenotype of glomerular and interstitial infiltrating cells in protocol renal allograft biopsies and histological diagnosis. Am J Transplant. 2007;7:2739–2747.
Scheepstra C, Bemelman FJ, van der Loos C, et al. B cells in cluster or in a scattered pattern do not correlate with clinical outcome of renal allograft rejection. Transplantation. 2008;86:772–778.
Cherukuri A, Salama AD, Mehta R, et al. Transitional B cell cytokines predict renal allograft outcomes. Sci Transl Med. 2021;13:eabe4929.
Miyajima M, Chase CM, Alessandrini A, et al. Early acceptance of renal allografts in mice is dependent on foxp3(+) cells. Am J Pathol. 2011;178:1635–1645.
Herman AE, Freeman GJ, Mathis D, et al. CD4+CD25+ T regulatory cells dependent on ICOS promote regulation of effector cells in the prediabetic lesion. J Exp Med. 2004;199:1479–1489.
Joshi NS, Akama-Garren EH, Lu Y, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity. 2015;43:579–590.
Peske JD, Thompson ED, Gemta L, et al. Effector lymphocyte-induced lymph node-like vasculature enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat Commun. 2015;6:7114.
Browning JL, Allaire N, Ngam-Ek A, et al. Lymphotoxin-beta receptor signaling is required for the homeostatic control of HEV differentiation and function. Immunity. 2005;23:539–550.
Hemmerich S, Bistrup A, Singer MS, et al. Sulfation of L-selectin ligands by an HEV-restricted sulfotransferase regulates lymphocyte homing to lymph nodes. Immunity. 2001;15:237–247.
Rodriguez AB, Parriott G, Engelhard VH. Tumor necrosis factor receptor regulation of peripheral node addressin biosynthetic components in tumor endothelial cells. Front Immunol. 2022;13:1009306.
Martinet L, Garrido I, Filleron T, et al. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 2011;71:5678–5687.
Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–565.
Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–555.
Petitprez F, de Reynies A, Keung EZ, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–560.
Datta RR, Schran S, Persa OD, et al. Post-transplant malignancies show reduced T-cell abundance and tertiary lymphoid structures as correlates of impaired cancer immunosurveillance. Clin Cancer Res. 2022;28:1712–1723.
Asleh R, Alnsasra H, Habermann TM, et al. Post-transplant lymphoproliferative disorder following cardiac transplantation. Front Cardiovasc Med. 2022;9:787975.
Zintzaras E, Voulgarelis M, Moutsopoulos HM. The risk of lymphoma development in autoimmune diseases: a meta-analysis. Arch Intern Med. 2005;165:2337–2344.
Mazzucchelli L, Blaser A, Kappeler A, et al. BCA-1 is highly expressed in Helicobacter pylori-induced mucosa-associated lymphoid tissue and gastric lymphoma. J Clin Invest. 1999;104:R49–R54.
Werner F, Wagner C, Simon M, et al. A standardized analysis of tertiary lymphoid structures in human melanoma: disease progression- and tumor site-associated changes with germinal center alteration. Front Immunol. 2021;12:675146.
Barmpoutis P, Di Capite M, Kayhanian H, et al. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cancer. PLoS One. 2021;16:e0256907.
Turvey SE, Swart E, Denis MC, et al. Noninvasive imaging of pancreatic inflammation and its reversal in type 1 diabetes. J Clin Invest. 2005;115:2454–2461.
Gaglia JL, Harisinghani M, Aganj I, et al. Noninvasive mapping of pancreatic inflammation in recent-onset type-1 diabetes patients. Proc Natl Acad Sci U S A. 2015;112:2139–2144.
Bentley KL, Stranford S, Liao S, et al. High endothelial venule reporter mice to probe regulation of lymph node vasculature. Adv Exp Med Biol. 2011;691:35–44.
Truman LA, Bentley KL, Smith EC, et al. ProxTom lymphatic vessel reporter mice reveal Prox1 expression in the adrenal medulla, megakaryocytes, and platelets. Am J Pathol. 2012;180:1715–1725.
Mohan JF, Kohler RH, Hill JA, et al. Imaging the emergence and natural progression of spontaneous autoimmune diabetes. Proc Natl Acad Sci U S A. 2017;114:E7776–E7785.
Fu W, Wojtkiewicz G, Weissleder R, et al. Early window of diabetes determinism in NOD mice, dependent on the complement receptor CRIg, identified by noninvasive imaging. Nat Immunol. 2012;13:361–368.
Jones GW, Bombardieri M, Greenhill CJ, et al. Interleukin-27 inhibits ectopic lymphoid-like structure development in early inflammatory arthritis. J Exp Med. 2015;212:1793–1802.
Gatumu MK, Skarstein K, Papandile A, et al. Blockade of lymphotoxin-beta receptor signaling reduces aspects of Sjogren’s syndrome in salivary glands of non-obese diabetic mice. Arthritis Res Ther. 2009;11:R24.
Masum MA, Ichii O, Elewa YHA, et al. Vasculature-associated lymphoid tissue: a unique tertiary lymphoid tissue correlates with renal lesions in lupus nephritis mouse model. Front Immunol. 2020;11:595672.
معلومات مُعتمدة: NIH; National Multiple Sclerosis Society
تواريخ الأحداث: Date Created: 20231102 Date Completed: 20240425 Latest Revision: 20240528
رمز التحديث: 20240528
مُعرف محوري في PubMed: PMC11042531
DOI: 10.1097/TP.0000000000004812
PMID: 37917987
قاعدة البيانات: MEDLINE
الوصف
تدمد:1534-6080
DOI:10.1097/TP.0000000000004812