1
|
Chambon M, Koenig A. NK Cells: Not Just Followers But Also Initiators of Chronic Vascular Rejection. Transpl Int 2024; 37:13318. [PMID: 39479216 PMCID: PMC11521863 DOI: 10.3389/ti.2024.13318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Chronic graft rejection represents a significant threat to long-term graft survival. Early diagnosis, understanding of the immunological mechanisms and appropriate therapeutic management are essential to improve graft survival and quality of life for transplant patients. Knowing which immune cells are responsible for chronic vascular rejection would allow us to provide effective and appropriate treatment for these patients. It is now widely accepted that natural killer (NK) cells play an important role in chronic vascular rejection. They can either initiate chronic vascular rejection by recognizing missing self on the graft or be recruited by donor-specific antibodies to destroy the graft during antibody-mediated rejection. Whatever the mechanisms of activation of NK cells, they need to be primed to become fully activated and damaging to the graft. A better understanding of the signaling pathways involved in NK cell priming and activation would pave the way for the development of new therapeutic strategies to cure chronic vascular rejection. This review examines the critical role of NK cells in the complex context of chronic vascular rejection.
Collapse
Affiliation(s)
- Mathilde Chambon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
2
|
Bailly E, Macedo C, Gu X, Hollingshead D, Bentlejewski C, Fong E, Morel PA, Randhawa P, Zeevi A, Lefaucheur C, Metes D. FCGR2C Q 13 and FCGR3A V 176 alleles jointly associate with worse natural killer cell-mediated antibody-dependent cellular cytotoxicity and microvascular inflammation in kidney allograft antibody-mediated rejection. Am J Transplant 2024:S1600-6135(24)00573-2. [PMID: 39332679 DOI: 10.1016/j.ajt.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/14/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024]
Abstract
Natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC) is a major mechanism of humoral allograft injury. FCGR3A V176/F176 polymorphism influences ADCC activity. Additionally, NK cell FcγRIIc expression, dictated by the Q13/STP13 polymorphism, was never investigated in kidney transplantation. To assess the clinical relevance of FCGR2C Q13/STP13 polymorphism in conjunction with FCGR3A V176/F176 polymorphism, 242 kidney transplant recipients were genotyped. NK cell Fc gamma receptor (FcγR) expression and ADCC activity were assessed. RNA sequencing was performed on kidney allograft biopsies to explore the presence of infiltrating FcγR+ NK cells. The FCGR2C Q13 allele was enriched in antibody-mediated rejection patients. FcγRIIc Q13+ NK cells had higher ADCC activity than FcγRIIc Q13- NK cells. In combination with the high-affinity FCGR3A V176 allele, Q13+V176+ NK cells were the most functionally potent. Q13+ was associated with worse microvascular inflammation and a higher risk of allograft loss. Among V176- patients, previously described in the literature as lower-risk patients, Q13+V176- showed a lower graft survival than Q13-V176- patients. In antibody-mediated rejection biopsies, FCGR2C transcripts were enriched and associated with ADCC-related transcripts. Our results suggest that FCGR2C Q13 in addition to FCGR3A V176 is a significant risk allele that may enhance NK cell-mediated ADCC and contribute to allograft injury and poor survival.
Collapse
Affiliation(s)
- Elodie Bailly
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; INSERM UMR-S976, Université Paris Cité, Paris, France.
| | - Camila Macedo
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xinyan Gu
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Hollingshead
- University of Pittsburgh Health Sciences Core Research Facilities, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carol Bentlejewski
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Erica Fong
- University of Pittsburgh Health Sciences Core Research Facilities, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Penelope A Morel
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Parmjeet Randhawa
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Adriana Zeevi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | - Diana Metes
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
3
|
Diebold M, Mayer KA, Hidalgo L, Kozakowski N, Budde K, Böhmig GA. Chronic Rejection After Kidney Transplantation. Transplantation 2024:00007890-990000000-00858. [PMID: 39192468 DOI: 10.1097/tp.0000000000005187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
In kidney transplantation, ongoing alloimmune processes-commonly triggered by HLA incompatibilities-can trigger chronic transplant rejection, affecting the microcirculation and the tubulointerstitium. Continuous inflammation may lead to progressive, irreversible graft injury, culminating in graft dysfunction and accelerated transplant failure. Numerous experimental and translational studies have delineated a complex interplay of different immune mechanisms driving rejection, with antibody-mediated rejection (AMR) being an extensively studied rejection variant. In microvascular inflammation, a hallmark lesion of AMR, natural killer (NK) cells have emerged as pivotal effector cells. Their essential role is supported by immunohistologic evidence, bulk and spatial transcriptomics, and functional genetics. Despite significant research efforts, a substantial unmet need for approved rejection therapies persists, with many trials yielding negative outcomes. However, several promising therapies are currently under investigation, including felzartamab, a monoclonal antibody targeting the surface molecule CD38, which is highly expressed in NK cells and antibody-producing plasma cells. In an exploratory phase 2 trial in late AMR, this compound has demonstrated potential in resolving molecular and morphologic rejection activity and injury, predominantly by targeting NK cell effector function. These findings inspire hope for effective treatments and emphasize the necessity of further pivotal trials focusing on chronic transplant rejection.
Collapse
Affiliation(s)
- Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Luis Hidalgo
- HLA Laboratory, Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Constantinides M, Robert N, Multrier C, Coënon L, Campos-Mora M, Jacquard C, Gao F, Zemiti S, Presumey J, Cartron G, Moreaux J, Villalba M. FCGR3A F158V alleles frequency differs in multiple myeloma patients from healthy population. Oncoimmunology 2024; 13:2388306. [PMID: 39175948 PMCID: PMC11340758 DOI: 10.1080/2162402x.2024.2388306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/28/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024] Open
Abstract
FCGR3A presents a single nucleotide polymorphism at location 158 (V/F), which affects its binding to the fragment crystallizable (Fc) of antibodies (Abs). FcγRIIIa-158 V allotype has the highest affinity and is associated with a better clinical response to IgG1 monoclonal Abs (mAb) treatment. We compared the allele frequency of FCGR3A-F158V polymorphism in cohorts of patients with B-cell lymphoproliferative disorders, including multiple myeloma (MM), monoclonal gammopathy of undetermined significance (MGUS), non-Hodgkin lymphoma (NHL), and B-cell chronic leukemia (B-CLL). FCGR3A-158F homozygous were enriched and tended to be in MM and MGUS patients, respectively; but neither in B-CLL nor in NHL patients. We identified a significantly lower concentration of CD8 T-cells and resting memory CD4 T-cells in MM patients bone marrow with the F/F genotype, associated with an increase in the macrophage percentage. In contrast, natural killer cells increased in V/V homozygous patients. This suggests a deregulation of the immune microenvironment in FCGR3A-F/F homozygous patients. However, we did not observe difference in response following treatment combining chemotherapy associated or not with daratumumab, an IgG1 mAb direct against CD38. Our findings suggest that FCGR3A F158V polymorphism can regulate the immune environment and affect the development of tumor plasma cells.
Collapse
Affiliation(s)
- Michaël Constantinides
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Caroline Multrier
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Loïs Coënon
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | | | - Carine Jacquard
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Fei Gao
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Sara Zemiti
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Jessy Presumey
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Jérome Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- IRMB, Univ Montpellier, INSERM, CNRS, CHRU de Montpellier, Montpellier, France
| |
Collapse
|
5
|
Diebold M, Vietzen H, Heinzel A, Haindl S, Herz CT, Mayer K, Doberer K, Kainz A, Faé I, Wenda S, Kühner LM, Berger SM, Puchhammer-Stöckl E, Kozakowski N, Schaub S, Halloran PF, Böhmig GA. Natural killer cell functional genetics and donor-specific antibody-triggered microvascular inflammation. Am J Transplant 2024; 24:743-754. [PMID: 38097018 DOI: 10.1016/j.ajt.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/28/2023] [Accepted: 12/02/2023] [Indexed: 12/31/2023]
Abstract
Antibody-mediated rejection (ABMR) is a leading cause of graft failure. Emerging evidence suggests a significant contribution of natural killer (NK) cells to microvascular inflammation (MVI). We investigated the influence of genetically determined NK cell functionality on ABMR development and activity. The study included 86 kidney transplant recipients subjected to systematic biopsies triggered by donor-specific antibody detection. We performed killer immunoglobulin-like receptor typing to predict missing self and genotyped polymorphisms determining NK cell functionality (FCGR3AV/F158 [rs396991], KLRC2wt/del, KLRK1HNK/LNK [rs1049174], rs9916629-C/T). Fifty patients had ABMR with considerable MVI and elevated NK cell transcripts. Missing self was not related to MVI. Only KLRC2wt/wt showed an association (MVI score: 2 [median; interquartile range: 0-3] vs 0 [0-1] in KLRC2wt/del recipients; P = .001) and remained significant in a proportional odds multivariable model (odds ratio, 7.84; 95% confidence interval, 2.37-30.47; P = .001). A sum score incorporating all polymorphisms and missing self did not outperform a score including only KLRC2 and FCGR3A variants, which were predictive in univariable analysis. NK cell genetics did not affect graft functional decline and survival. In conclusion, a functional KLRC2 polymorphism emerged as an independent determinant of ABMR activity, without a considerable contribution of missing self and other NK cell gene polymorphisms.
Collapse
Affiliation(s)
- Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Susanne Haindl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Carsten T Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alexander Kainz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ingrid Faé
- Department of Blood Group Serology and Transfusion Medicine, Medical University Vienna, Vienna, Austria
| | - Sabine Wenda
- Department of Blood Group Serology and Transfusion Medicine, Medical University Vienna, Vienna, Austria
| | - Laura M Kühner
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Sarah M Berger
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | | | - Stefan Schaub
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre, ATAGC, University of Alberta, Edmonton, Alberta, Canada
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Pang Q, Chen L, An C, Zhou J, Xiao H. Single-cell and bulk RNA sequencing highlights the role of M1-like infiltrating macrophages in antibody-mediated rejection after kidney transplantation. Heliyon 2024; 10:e27865. [PMID: 38524599 PMCID: PMC10958716 DOI: 10.1016/j.heliyon.2024.e27865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Background Antibody-mediated rejection (ABMR) significantly affects transplanted kidney survival, yet the macrophage phenotype, ontogeny, and mechanisms in ABMR remain unclear. Method We analyzed post-transplant sequencing and clinical data from GEO and ArrayExpress. Using dimensionality reduction and clustering on scRNA-seq data, we identified macrophage subpopulations and compared their infiltration in ABMR and non-rejection cases. Cibersort quantified these subpopulations in bulk samples. Cellchat, SCENIC, monocle2, and monocle3 helped explore intercellular interactions, predict transcription factors, and simulate differentiation of cell subsets. The Scissor method linked macrophage subgroups with transplant prognosis. Furthermore, hdWGCNA, nichnet, and lasso regression identified key genes associated with core transcription factors in selected macrophages, validated by external datasets. Results Six macrophage subgroups were identified in five post-transplant kidney biopsies. M1-like infiltrating macrophages, prevalent in ABMR, correlated with pathological injury severity. MIF acted as a primary intercellular signal in these macrophages. STAT1 regulated monocyte-to-M1-like phenotype transformation, impacting transplant prognosis via the IFNγ pathway. The prognostic models built on the upstream and downstream genes of STAT1 effectively predicted transplant survival. The TLR4-STAT1-PARP9 axis may regulate the pro-inflammatory phenotype of M1-like infiltrating macrophages, identifying PARP9 as a potential target for mitigating ABMR inflammation. Conclusion Our study delineates the macrophage landscape in ABMR post-kidney transplantation, underscoring the detrimental impact of M1-like infiltrating macrophages on ABMR pathology and prognosis.
Collapse
Affiliation(s)
- Qidan Pang
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Liang Chen
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Changyong An
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Juan Zhou
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Hanyu Xiao
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| |
Collapse
|
7
|
Conley HE, He MM, Easterhoff D, Kirshner HF, Cocklin SL, Meyer J, Hoxie T, Berry M, Bradley T, Tolbert WD, Pazgier M, Tomaras GD, Schmitz JE, Moody MA, Wiehe K, Pollara J. Defining genetic diversity of rhesus macaque Fcγ receptors with long-read RNA sequencing. Front Immunol 2024; 14:1306292. [PMID: 38264644 PMCID: PMC10803544 DOI: 10.3389/fimmu.2023.1306292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Fcγ receptors (FcγRs) are membrane-bound glycoproteins that bind to the fragment crystallizable (Fc) constant regions of IgG antibodies. Interactions between IgG immune complexes and FcγRs can initiate signal transduction that mediates important components of the immune response including activation of immune cells for clearance of opsonized pathogens or infected host cells. In humans, many studies have identified associations between FcγR gene polymorphisms and risk of infection, or progression of disease, suggesting a gene-level impact on FcγR-dependent immune responses. Rhesus macaques are an important translational model for most human health interventions, yet little is known about the breadth of rhesus macaque FcγR genetic diversity. This lack of knowledge prevents evaluation of the impact of FcγR polymorphisms on outcomes of preclinical studies performed in rhesus macaques. In this study we used long-read RNA sequencing to define the genetic diversity of FcγRs in 206 Indian-origin Rhesus macaques, Macaca mulatta. We describe the frequency of single nucleotide polymorphisms, insertions, deletions, frame-shift mutations, and isoforms. We also index the identified diversity using predicted and known rhesus macaque FcγR and Fc-FcγR structures. Future studies that define the functional significance of this genetic diversity will facilitate a better understanding of the correlation between human and macaque FcγR biology that is needed for effective translation of studies with antibody-mediated outcomes performed in rhesus macaques.
Collapse
Affiliation(s)
- Haleigh E. Conley
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Max M. He
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - David Easterhoff
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Hélène Fradin Kirshner
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Sarah L. Cocklin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jacob Meyer
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Taylor Hoxie
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Todd Bradley
- Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, MO, United States
| | - William D. Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgia D. Tomaras
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Joern E. Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Michael Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Justin Pollara
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
8
|
Betjes MGH, Kal-van Gestel J, Roodnat JI, de Weerd AE. The Incidence of Antibody-Mediated Rejection Is Age-Related, Plateaus Late After Kidney Transplantation, and Contributes Little to Graft Loss in the Older Recipients. Transpl Int 2023; 36:11751. [PMID: 38188697 PMCID: PMC10768842 DOI: 10.3389/ti.2023.11751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
It is not known whether antibody-mediated rejection (ABMR) is age-related, whether it plateaus late after transplantation, and to what extent it contributes to graft loss in older recipients. Patients transplanted between 2010 and 2015 (n = 1,054) in a single center had regular follow-up until January 2023. Recipients were divided into age groups at transplantation: 18-39 years ("young"), 40-55 years ("middle age"), and >55 years ("elderly"). Ten years after transplantation the cumulative % of recipients with ABMR was 17% in young, 15% in middle age, and 12% in elderly recipients (p < 0.001). The cumulative incidence of ABMR increased over time and plateaued 8-10 years after transplantation. In the elderly, with a median follow-up of 7.5 years, on average 30% of the recipients with ABMR died with a functional graft and ABMR contributed only 4% to overall graft loss in this group. These results were cross-validated in a cohort of recipients with >15 years follow-up. Multivariate cox-regression analysis showed that increasing recipient age was independently associated with decreasing risk for ABMR. In conclusion, the cumulative risk for ABMR is age-dependent, plateaus late after transplantation, and contributes little to overall graft loss in older recipients.
Collapse
Affiliation(s)
- Michiel G. H. Betjes
- Rotterdam Transplantation Institute, Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
9
|
Asmaa C, Rachida R, Asma D, Louiza K, Souad C, Leila B, Ali B, Messaoud S, Dalila K, Fethi M, Nawel S, Malika A, Bachira M, Nabila A, Chafia TB, Habiba AAB. TNF-α -308A/G SNP association with kidney allograft rejection in Algerian population: A retrospective case-control study. Transpl Immunol 2023; 81:101927. [PMID: 37683737 DOI: 10.1016/j.trim.2023.101927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
No consensus has been reached regarding the association beween the -308A/G single nucleotide polymorphism (SNP) in the tumor necrosis factor-α gene (TNFA) and kidney allograft rejection (KAR). Our retrospective case-control study aimed to assess the association of the SNP with KAR in Algerian patients who underwent kidney transplantation. The study enrolled 313 Algerian patients: 58 kidney-transplant recipients without rejection events (PWoR); 58 kidney-transplant recipients with at least one rejection event, with or without graft loss (PWR); and 197 healthy individuals (HI). The TNFA -308A/G SNP was genotyped using a real-time polymerase chain reaction. The results demonstrated that, the frequencies of TNFA -308A allele and AA genotype were higher in the PWR than in the HI groups (p = 0.001, OR = 2.26, CI = 1.33-3.77 and p = 0.0004, OR = 5.53, CI-1.89-16.6, respectively). Furthermore, the frequencies were higher among the PWR than among the PWoR groups (p = 0.001, OR = 3.29, CI = 1.56-7.21 and p = 0.0006, OR = 28.26, CI = 1.62-493.2, respectively), particularly among PWR patients with de novo anti-human leukocyte antigens (HLA) antibodies (PG-a-HLA-Ab). However, the frequency of TNFA -308G allele was lower in the PWR group than in the PWoR group (p = 0.001, OR = 0.3, CI = 0.1-0.64) and the HI group (p = 0.001, OR = 0.44, CI = 0.27-0.44). Our results suggest an association of the TNFA -308A/G alleles with KAR in Algerian patients who underwent kidney transplantation. Carriers of TNFA -308A allele who have PG-a-HLA-Ab might have a higher risk, whereas TNFA -308G allele carriers could have a lower risk of KAR. Thus, therapeutic strategies can be adapted to minimize KAR risk in patients who have a genetic proclivity for increased pro-inflammatory TNF-α activity.
Collapse
Affiliation(s)
- Chebine Asmaa
- Cytokine and NO Synthase Team, Cellular and Molecular Biology Laboratory, Biological Sciences Faculty, University of Sciences and Technologies Houari Boumedienne (LBCM-FSB-USTHB), Algiers, Algeria
| | - Raache Rachida
- Cytokine and NO Synthase Team, Cellular and Molecular Biology Laboratory, Biological Sciences Faculty, University of Sciences and Technologies Houari Boumedienne (LBCM-FSB-USTHB), Algiers, Algeria
| | - Djendi Asma
- Transplantation and Immunogenetics Laboratory, Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Kaci Louiza
- Histological Analysis Laboratory, Algiers, Algeria
| | - Chelghoum Souad
- Department of Nephrology and Transplantation, Nafissa Hamoud Hospital, Algiers, Algeria
| | - Belkacemi Leila
- Department of Nephrology and Transplantation, Nafissa Hamoud Hospital, Algiers, Algeria
| | - Benziane Ali
- Department of Nephrology and Transplantation, Mohamed Amine Debbaghine Hospital, Algiers, Algeria
| | - Saidani Messaoud
- Department of Nephrology and Transplantation, Asaad Hassani Hospital, Algiers, Algeria
| | - Khemri Dalila
- Department of Nephrology and Transplantation, Mustapha Pacha Hospital, Algiers, Algeria
| | - Meҫabih Fethi
- Transplantation and Immunogenetics Laboratory, Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Salhi Nawel
- Transplantation and Immunogenetics Laboratory, Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Akachouche Malika
- Transplantation and Immunogenetics Laboratory, Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Mechti Bachira
- Transplantation and Immunogenetics Laboratory, Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Attal Nabila
- Immunology Department, Pasteur Institute of Algeria, Algiers, Algeria
| | - Touil-Boukoffa Chafia
- Cytokine and NO Synthase Team, Cellular and Molecular Biology Laboratory, Biological Sciences Faculty, University of Sciences and Technologies Houari Boumedienne (LBCM-FSB-USTHB), Algiers, Algeria
| | | |
Collapse
|
10
|
Franco-Acevedo A, Pathoulas CL, Murphy PA, Valenzuela NM. The Transplant Bellwether: Endothelial Cells in Antibody-Mediated Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1276-1285. [PMID: 37844279 PMCID: PMC10593495 DOI: 10.4049/jimmunol.2300363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/22/2023] [Indexed: 10/18/2023]
Abstract
Ab-mediated rejection of organ transplants remains a stubborn, frequent problem affecting patient quality of life, graft function, and grant survival, and for which few efficacious therapies currently exist. Although the field has gained considerable knowledge over the last two decades on how anti-HLA Abs cause acute tissue injury and promote inflammation, there has been a gap in linking these effects with the chronic inflammation, vascular remodeling, and persistent alloimmunity that leads to deterioration of graft function over the long term. This review will discuss new data emerging over the last 5 y that provide clues into how ongoing Ab-endothelial cell interactions may shape vascular fate and propagate alloimmunity in organ transplants.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA
| | | | - Patrick A Murphy
- Center for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
11
|
Betjes MGH, De Weerd A. Lowering maintenance immune suppression in elderly kidney transplant recipients; connecting the immunological and clinical dots. Front Med (Lausanne) 2023; 10:1215167. [PMID: 37502354 PMCID: PMC10368955 DOI: 10.3389/fmed.2023.1215167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/09/2023] [Indexed: 07/29/2023] Open
Abstract
The management of long-term immune suppressive medication in kidney transplant recipients is a poorly explored field in the area of transplant medicine. In particular, older recipients are at an increased risk for side effects and have an exponentially increased risk of infection-related death. In contrast, an aged immune system decreases the risk of acute T-cell-mediated rejection in older recipients. Recent advances in alloimmunity research have shown a rapid and substantial decline in polyfunctional, high-risk CD4+ T cells post-transplantation. This lowers the direct alloreactivity responsible for T-cell-mediated rejection, also known as donor-specific hyporesponsiveness. Chronic antibody-mediated rejection (c-aABMR) is the most frequent cause of kidney graft loss in the long term. However, in older adults, c-aABMR as a cause of graft loss is outnumbered by death with a functioning graft. In addition, DSA development and a diagnosis of c-aABMR plateau ~10 years after transplantation, resulting in a very low risk for rejection thereafter. The intensity of immune suppression regimes could likely be reduced accordingly, but trials in this area are scarce. Tacrolimus monotherapy for 1 year after transplantation seems feasible in older kidney transplant recipients with standard immunological risk, showing the expected benefits of fewer infections and better vaccination responses.
Collapse
|
12
|
Zhang Q, Ouyang Q, Li W, Chiu J, Yan M, Lu YS, Sun S, Li H, Du Y, Wang X, Sun T, Yin Y, Wang H, Ye F, Shen K, Wang J, Pan Y, Wang S, Yang J, Wu X, Dai MS, Cheng J, Teng Y, Su F, Wu X, He J, Fu P, Yang L, Xin Y, Wang X, Jiang Z. Efficacy and safety of margetuximab plus chemotherapy vs. trastuzumab plus chemotherapy in Chinese patients with pretreated HER2-positive advanced metastatic breast cancer: results from a randomized, open-label, multicenter, phase II bridging study. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2022; 3:31. [PMID: 38751523 PMCID: PMC11093073 DOI: 10.21037/tbcr-22-35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/15/2022] [Indexed: 05/18/2024]
Abstract
Background Trastuzumab is the recommended first-line treatment for human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC) patients in China, but therapeutic resistance to trastuzumab and other early-line treatments is common and late-line treatment options are limited. Derived from the same murine precursor antibody, margetuximab has enhanced anti-tumor activity compared with trastuzumab and may be an effective late-line treatment. However, data regarding the use of margetuximab in pre-treated Chinese patients are scarce. This study aimed to evaluate the efficacy and safety of margetuximab plus chemotherapy vs. trastuzumab plus chemotherapy in Chinese patients, and to determine whether the results are consistent with the clinical benefit of margetuximab observed in the pivotal global phase III study. Methods In this randomized, open-label, multicenter, phase II bridging study, eligible Chinese patients pretreated with ≥2 lines of anti-HER2 therapies were randomized 1:1 by stratified block randomization to margetuximab (15 mg/kg over at least 120 minutes) or trastuzumab (6 mg/kg over at least 30 minutes), each administered intravenously once every 21-day cycle and plus chemotherapy. Disease assessment was conducted once every two treatment cycles (6 weeks ± 7 days). The primary endpoint was progression-free survival (PFS) by blinded independent central review (BICR). Secondary endpoints included overall survival (OS), investigator-assessed PFS, objective response rate (ORR), duration of response (DoR), clinical benefit rate (CBR), and the incidence and severity of treatment-emergent adverse events (TEAEs). Results Between February 4, 2020 and February 23, 2021, 123 patients were randomized to the margetuximab (n=62) and trastuzumab (n=61) arms. Among them, 15 and 7 patients, respectively, were still on study treatment as of data cut-off (September 3, 2021). Overall, 99.2% were female, median age was 53 years old. All patients were pretreated with trastuzumab, and 83.7% and 25.2%, respectively, were pretreated with tyrosine kinase inhibitors (TKIs) and pertuzumab. Baseline characteristics were numerically balanced between arms. BICR-assessed median PFS (mPFS) was 5.5 months in the margetuximab arm and 4.1 months in the trastuzumab arm, with a hazard ratio (HR) of 0.69 [95% confidence interval (CI): 0.43-1.12], which met the consistency criterion (HR <0.88) for bridging success. Median investigator-assessed PFS was 5.5 months in the margetuximab arm and 4.0 months in the trastuzumab arm (HR =0.63; 95% CI: 0.41-0.96). Median OS (mOS) was not yet reached. Both ORR and CBR were greater in the margetuximab arm (25.5% vs. 12.5%; 32.7% vs. 14.3%). Safety results were numerically comparable between the two arms. Anti-HER2 treatment-related infusion-related reactions (IRRs) were more common in the margetuximab arm than in the trastuzumab arm (12.9% vs. 1.7%). All IRRs could be resolved. Conclusions Margetuximab was effective and well-tolerated in this study, supporting its clinical use in pretreated HER2-positive MBC patients in China. Trial Registration ClinicalTrials.gov Identifier: NCT04262804.
Collapse
Affiliation(s)
- Qingyuan Zhang
- Department of Breast Cancer, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Quchang Ouyang
- Department of Medical Oncology, Hunan Cancer Hospital, Changsha, China
| | - Wei Li
- Department of Medical Oncology, The First Hospital of Jilin University, Changchun, China
| | - Joanne Chiu
- Division of Hematology and Medical Oncology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Min Yan
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Yen-Shen Lu
- Department of Medical Oncology, Taiwan University Hospital, Taipei
| | - Sanyuan Sun
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou, China
| | - Huiping Li
- Department of Breast Cancer, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yingying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xujuan Wang
- Department of Breast Cancer, The Second People’s Hospital of Neijiang, Neijiang, China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yongmei Yin
- Department of Oncology, Jiangsu Province Hospital, Nanjing, China
| | - Haibo Wang
- Department of Breast Cancer, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Feng Ye
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, China
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Kunwei Shen
- Department of Breast Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingfen Wang
- Department of Breast Cancer, Linyi Cancer Hospital, Linyi, China
| | - Yueyin Pan
- Department of Medical Oncology/Chemotherapy, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, China
| | - Shusen Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jin Yang
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaohong Wu
- Department of Medical Oncology, The Affiliated Hospital of Jiangnan University (The Fourth People’s Hospital in Wuxi City), Wuxi, China
| | - Ming-Shen Dai
- Division of Oncology, Department of Medicine, Tri-Service General Hospital, Taipei
| | - Jing Cheng
- Department of Breast Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuee Teng
- Department of Breast Cancer, The First Hospital of China Medical University, Shenyang, China
| | - Fang Su
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xinhong Wu
- Department of Breast Oncology, Hubei Cancer Hospital, Wuhan, China
| | - Jingdong He
- Department of Medical Oncology, Huai’an First People’s Hospital, Huai’an, China
| | - Peifen Fu
- Department of Breast Cancer, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lulu Yang
- Department of Research & Development, Zai Lab (Shanghai) Co., Ltd., Shanghai, China
| | - Yuan Xin
- Department of Research & Development, Zai Lab (Shanghai) Co., Ltd., Shanghai, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Zefei Jiang
- Department of Breast Cancer, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
Delpire B, Van Loon E, Naesens M. The Role of Fc Gamma Receptors in Antibody-Mediated Rejection of Kidney Transplants. Transpl Int 2022; 35:10465. [PMID: 35935272 PMCID: PMC9346079 DOI: 10.3389/ti.2022.10465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
For the past decades, complement activation and complement-mediated destruction of allograft cells were considered to play a central role in anti-HLA antibody-mediated rejection (AMR) of kidney transplants. However, also complement-independent mechanisms are relevant in the downstream immune activation induced by donor-specific antibodies, such as Fc-gamma receptor (FcγR)-mediated direct cellular activation. This article reviews the literature regarding FcγR involvement in AMR, and the potential contribution of FcγR gene polymorphisms to the risk for antibody mediated rejection of kidney transplants. There is large heterogeneity between the studies, both in the definition of the clinical phenotypes and in the technical aspects. The study populations were generally quite small, except for two larger study cohorts, which obviates drawing firm conclusions regarding the associations between AMR and specific FcγR polymorphisms. Although FcγR are central in the pathophysiology of AMR, it remains difficult to identify genetic risk factors for AMR in the recipient’s genome, independent of clinical risk factors, independent of the donor-recipient genetic mismatch, and in the presence of powerful immunosuppressive agents. There is a need for larger, multi-center studies with standardised methods and endpoints to identify potentially relevant FcγR gene polymorphisms that represent an increased risk for AMR after kidney transplantation.
Collapse
Affiliation(s)
- Boris Delpire
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Naesens
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| |
Collapse
|
14
|
Betjes MGH, Roelen DL, van Agteren M, Kal-van Gestel J. Causes of Kidney Graft Failure in a Cohort of Recipients With a Very Long-Time Follow-Up After Transplantation. Front Med (Lausanne) 2022; 9:842419. [PMID: 35733857 PMCID: PMC9207199 DOI: 10.3389/fmed.2022.842419] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/05/2022] [Indexed: 01/03/2023] Open
Abstract
Background Biopsy-proven causes of graft loss many years after kidney transplantation are scarcely documented. Methods Patients transplanted between 1995 and 2005 (n = 737) in a single center were followed on a regular basis until 2021. The recipients were divided according to age at transplantation into 3 groups; 18–39 years (young), 40–55 years (middle age), and older than 55 years (elderly). For cause biopsies of renal transplants were clustered into the categories, rejection, IFTA, return original disease, and diagnosis of de novo kidney disease. Results Rejection was the main cause of graft failure censored for death at every time period after transplantation. The incidence of T cell-mediated rejection (TCMR) became rare 6 years after transplantation while the cumulative incidence of antibody-mediated rejection (ABMR) increased over time (1.1% per year). ABMR was not diagnosed anymore beyond 15 years of follow-up in recipients without pre-transplant donor-specific antibodies (DSA). An episode of TCMR was associated with an increased incidence of ABMR diagnosis in the short-term but did not increase the overall incidence of AMBR not in the long-term. Death as a cause of graft failure was an important competitive risk factor long after transplantation and resulted in a significantly lower frequency of rejection-related graft loss in the elderly group (11 vs. 23% in the young group at 15 year follow-up). Conclusion Rejection is a major cause of graft loss but recipient’s age, time after transplantation, and the presence of DSA before transplantation determine the relative contribution to overall graft loss and the type of rejection involved.
Collapse
Affiliation(s)
- Michiel G. H. Betjes
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
- *Correspondence: Michiel G. H. Betjes, ; orcid.org/0000-0001-9435-6208
| | - Dave L. Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Madelon van Agteren
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
| | - Judith Kal-van Gestel
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
| |
Collapse
|
15
|
Jethwani P, Rao A, Bow L, Menon MC. Donor–Recipient Non-HLA Variants, Mismatches and Renal Allograft Outcomes: Evolving Paradigms. Front Immunol 2022; 13:822353. [PMID: 35432337 PMCID: PMC9012490 DOI: 10.3389/fimmu.2022.822353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/03/2022] [Indexed: 12/22/2022] Open
Abstract
Despite significant improvement in the rates of acute allograft rejection, proportionate improvements in kidney allograft longevity have not been realized, and are a source of intense research efforts. Emerging translational data and natural history studies suggest a role for anti-donor immune mechanisms in a majority of cases of allograft loss without patient death, even when overt evidence of acute rejection is not identified. At the level of the donor and recipient genome, differences in highly polymorphic HLA genes are routinely evaluated between donor and recipient pairs as part of organ allocation process, and utilized for patient-tailored induction and maintenance immunosuppression. However, a growing body of data have characterized specific variants in donor and recipient genes, outside of HLA loci, that induce phenotypic changes in donor organs or the recipient immune system, impacting transplant outcomes. Newer mechanisms for “mismatches” in these non-HLA loci have also been proposed during donor–recipient genome interactions with transplantation. Here, we review important recent data evaluating the role of non-HLA genetic loci and genome-wide donor-recipient mismatches in kidney allograft outcomes.
Collapse
Affiliation(s)
- Priyanka Jethwani
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Arundati Rao
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Laurine Bow
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Madhav C. Menon
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- *Correspondence: Madhav C. Menon,
| |
Collapse
|
16
|
Vietzen H, Döhler B, Tran TH, Süsal C, Halloran PF, Eskandary F, Herz CT, Mayer KA, Kozakowski N, Wahrmann M, Ely S, Haindl S, Puchhammer-Stöckl E, Böhmig GA. Deletion of the Natural Killer Cell Receptor NKG2C Encoding KLR2C Gene and Kidney Transplant Outcome. Front Immunol 2022; 13:829228. [PMID: 35401541 PMCID: PMC8987017 DOI: 10.3389/fimmu.2022.829228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells may contribute to antibody-mediated rejection (ABMR) of renal allografts. The role of distinct NK cell subsets in this specific context, such as NK cells expressing the activating receptor NKG2C, is unknown. Our aim was to investigate whether KLRC2 gene deletion variants which determine NKG2C expression affect the pathogenicity of donor-specific antibodies (DSA) and, if so, influence long-term graft survival. We genotyped the KLRC2wt/del variants for two distinct kidney transplant cohorts, (i) a cross-sectional cohort of 86 recipients who, on the basis of a positive post-transplant DSA result, all underwent allograft biopsies, and (ii) 1,860 recipients of a deceased donor renal allograft randomly selected from the Collaborative Transplant Study (CTS) database. In the DSA+ patient cohort, KLRC2wt/wt (80%) was associated with antibody-mediated rejection (ABMR; 65% versus 29% among KLRC2wt/del subjects; P=0.012), microvascular inflammation [MVI; median g+ptc score: 2 (interquartile range: 0-4) versus 0 (0-1), P=0.002], a molecular classifier of ABMR [0.41 (0.14-0.72) versus 0.10 (0.07-0.27), P=0.001], and elevated NK cell-related transcripts (P=0.017). In combined analyses of KLRC2 variants and a functional polymorphism in the Fc gamma receptor IIIA gene (FCGR3A-V/F158), ABMR rates and activity gradually increased with the number of risk genotypes. In DSA+ and CTS cohorts, however, the KLRC2wt/wt variant did not impact long-term death-censored graft survival, also when combined with the FCGR3A-V158 risk variant. KLRC2wt/wt may be associated with DSA-triggered MVI and ABMR-associated gene expression patterns, but the findings observed in a highly selected cohort of DSA+ patients did not translate into meaningful graft survival differences in a large multicenter kidney transplant cohort not selected for HLA sensitization.
Collapse
Affiliation(s)
- Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Bernd Döhler
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thuong Hien Tran
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Caner Süsal
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Transplant Immunology Research Center of Excellence, Koç Üniversitesi, Istanbul, Turkey
| | - Philip F. Halloran
- Alberta Transplant Applied Genomics Centre (ATAGC), University of Alberta, Edmonton, AB, Canada
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Carsten T. Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina A. Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sarah Ely
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Susanne Haindl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Puchhammer-Stöckl
- Center for Virology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Georg A. Böhmig, ; Elisabeth Puchhammer-Stöckl,
| | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- *Correspondence: Georg A. Böhmig, ; Elisabeth Puchhammer-Stöckl,
| |
Collapse
|
17
|
Wahrmann M, Döhler B, Arnold ML, Scherer S, Mayer KA, Haindl S, Haslacher H, Böhmig GA, Süsal C. Functional Fc Gamma Receptor Gene Polymorphisms and Long-Term Kidney Allograft Survival. Front Immunol 2021; 12:724331. [PMID: 34497614 PMCID: PMC8420807 DOI: 10.3389/fimmu.2021.724331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
The functional Fc gamma receptor (FcγR) IIIA polymorphism FCGR3A-V/F158 was earlier suggested to determine the potential of donor-specific HLA antibodies to trigger microcirculation inflammation, a key lesion of antibody-mediated renal allograft rejection. Associations with long-term transplant outcomes, however, have not been evaluated to date. To clarify the impact of FCGR3A-V/F158 polymorphism on kidney transplant survival, we genotyped a cohort of 1,940 recipient/donor pairs. Analyzing 10-year death-censored allograft survival, we found no significant differences in relation to FCGR3A-V/F158. There was also no independent survival effect in a multivariable Cox model. Similarly, functional polymorphisms in two other activating FcγR, FCGR2A-H/R131 (FcγRIIA) and FCGR3B-NA1/NA2 (FcγRIIIB), were not associated with outcome. There were also no significant survival differences among patient subgroups at increased risk of rejection-related injury, such as pre-sensitized recipients (> 0% panel reactivity; n = 438) or recipients treated for rejection within the first year after transplantation (n = 229). Our study results suggest that the earlier shown association of FcγR polymorphism with microcirculation inflammation may not be strong enough to exert a meaningful effect on graft survival.
Collapse
Affiliation(s)
- Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bernd Döhler
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marie-Luise Arnold
- Department of Internal Medicine 3, Institute for Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sabine Scherer
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Susanne Haindl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Caner Süsal
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|