1
|
Nankivell BJ. Evolving thresholds for the diagnosis of acute T cell mediated rejection. Curr Opin Nephrol Hypertens 2025; 34:212-217. [PMID: 40104932 DOI: 10.1097/mnh.0000000000001072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW The Banff schema uses combinations of pathological lesions at predefined thresholds to diagnose of T cell rejection (TCMR) and grade its severity. Constant definitional changes have caused confusion among clinicians and pathologists. This review describes the evolution of lesion definitions and the rationale for the minimal thresholds. RECENT FINDINGS The minimal diagnostic threshold for borderline TCMR has been reset to original Banff i1/t1, where isolated tubulitis is now excluded. Arteritis can be mediated by either Grade II TCMR or caused by donor specific antibody as antibody-mediated vascular rejection. The conservative threshold for chronic active TCMR diagnosis uses moderate total and scarred inflammation with tubulitis has been challenged by recent longitudinal data to suggest lower thresholds including i-IFTA=1 as clinically relevant. SUMMARY Minor changes in the threshold ruleset can cause substantial alterations in the final pathological diagnoses. While minimal thresholds for borderline and active TCMR have now stabilized, future changes are likely for chronic active TCMR pending confirmatory research.
Collapse
|
2
|
Torres Sánchez MJ, Ruiz Fuentes MC, Clavero García E, Rísquez Chica N, Espinoza Muñoz K, Espigares Huete MJ, Caba Molina M, Osuna A, Wangensteen R. Hydroxyproline in Urine Microvesicles as a Biomarker of Fibrosis in the Renal Transplant Patient. Biomedicines 2024; 12:2836. [PMID: 39767742 PMCID: PMC11673537 DOI: 10.3390/biomedicines12122836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Interstitial fibrosis/tubular atrophy in kidney transplantation is an unspecific lesion induced by immune and non-immune factors, which determines the progression of chronic kidney disease. Hydroxyproline is an imino acid that is part of the molecule of collagen. The aim of this study was to assess hydroxyproline in urine microvesicles as a marker of fibrosis in the renal transplant patient. Patients and Methods: An observational cross-sectional study was conducted on 46 renal transplant patients who had undergone renal biopsy with diagnostic intention, as well as 19 healthy controls. Clinical, histological, and laboratory variables were collected at the time of marker determination and renal function was analyzed 2 years later. Hydroxyproline was measured in urine microvesicles. Results: Renal transplant patients showed a higher microvesicular concentration of hydroxyproline compared to the control group, with the following medians (interquartile range (IQR)): 28.024 (5.53) ng/mL vs. 2.51 (1.16) ng/mL, p < 0.001. In the transplanted patients, patients in whom biopsy showed some score of total cortical parenchymal inflammation (ti) displayed a significantly higher concentration of hydroxyproline in urine microvesicles than those patients who did not score for cortical parenchymal inflammation (29.91 ± 2.797 ng/mL vs. 22.72 ± 8.697 ng/mL, p = 0.034). No significant correlation was observed between urinary markers and serum creatinine, calcium, and parathyroid hormone (PTH). Conclusions: The concentration of hydroxyproline in urinary microvesicles increased in renal transplant patients relative to healthy controls. Hydroxyproline in urinary microvesicles is a marker of chronic renal inflammation in transplanted patients, and further studies are required to confirm this finding in other pathologies, as well as the association with fibrosis and the evolution of renal function.
Collapse
Affiliation(s)
- María José Torres Sánchez
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
| | - María Carmen Ruiz Fuentes
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
- Department of Medicine, University of Granada, 18071 Granada, Spain;
| | - Elena Clavero García
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
| | - Noelia Rísquez Chica
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
| | - Karla Espinoza Muñoz
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
| | - María José Espigares Huete
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain; (M.J.T.S.); (E.C.G.); (N.R.C.); (K.E.M.); (M.J.E.H.)
| | - Mercedes Caba Molina
- Department of Pathological Anatomy, Provincial Unit of Pathological Anatomy of Granada (UPIGAP), Instituto de Investigación Biosanitaria, Ibs Granada, University of Granada, 18071 Granada, Spain;
| | - Antonio Osuna
- Department of Medicine, University of Granada, 18071 Granada, Spain;
| | - Rosemary Wangensteen
- Area of Physiology, Department of Health Sciencies, University of Jaen, 23071 Jaen, Spain;
| |
Collapse
|
3
|
Mulley WR, Hughes PD, Collins MG, Pilmore HL, Clayton PA, Wyld ML, Lee D, van der Jeugd J, Fernando SC, Kuo SF, Tan S, Jahan S, Lim WH. Defining causes of death-censored kidney allograft failure: A 5-year multicentre ANZDATA and clinical cross-sectional study. Nephrology (Carlton) 2024; 29:930-940. [PMID: 39349052 PMCID: PMC11579561 DOI: 10.1111/nep.14397] [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: 04/24/2024] [Revised: 08/08/2024] [Accepted: 09/14/2024] [Indexed: 10/02/2024]
Abstract
AIM Determining specific causes of allograft failure allows a focus on understanding and treating these conditions. Previous studies highlight chronic antibody-mediated rejection as a leading cause of late allograft failure. We sought to define causes of allograft failure in a large cohort of kidney transplant recipients across multiple centres in Australia and New Zealand, including cases previously attributed to chronic allograft nephropathy (CAN). METHODS All death-censored allograft failures at 9 participating centres between 1 January 2014 to 31 December 2018 were included. Available clinical and biopsy data were reviewed and the "most likely" cause assigned. RESULTS There were 642 death-censored allograft failures in the study period. Of these, 495 (77.1%) had an informative biopsy performed a median of 13.4 months (IQR 2.5-39.1 months) prior to allograft failure. Rejection of any type was the leading cause of allograft failure (47.5%), comprised chiefly of chronic antibody-mediated rejection (37.4%) and chronic T-cell mediated rejection (6.4%). Other leading causes were undifferentiated interstitial fibrosis and tubular atrophy (10.8%), late medical and surgical complications (8.1%) and recurrent or de novo glomerulonephritis (7.0%). Polyoma viral nephropathy and calcineurin inhibitor toxicity each contributed to <2%. Causes of allograft failure previously attributed to CAN (n = 419, 65.3%) had a similar distribution to the overall cohort, with 43.9% attributed to chronic antibody-mediated rejection. CONCLUSION To prolong allograft survival, improved strategies are needed to curtail alloimmune responses. Greater understanding of the causes of undifferentiated interstitial fibrosis and tubular atrophy and potential treatments would also be of considerable benefit.
Collapse
Affiliation(s)
- William R. Mulley
- Department of NephrologyMonash Medical CentreClaytonVictoriaAustralia
- Centre for Inflammatory Diseases, Department of MedicineMonash UniversityClaytonVictoriaAustralia
| | - Peter D. Hughes
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of MedicineThe University of MelbourneMelbourneVictoriaAustralia
| | - Michael G. Collins
- Department of Renal MedicineAuckland City HospitalAucklandNew Zealand
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideAustralia
| | - Helen L. Pilmore
- Department of Renal MedicineAuckland City HospitalAucklandNew Zealand
- Department of MedicineUniversity of AucklandAucklandNew Zealand
| | - Philip A. Clayton
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideAustralia
- Australia & New Zealand Dialysis and Transplant (ANZDATA) RegistryAdelaideAustralia
| | - Melanie L. Wyld
- Department of Renal and Transplant MedicineWestmead HospitalWestmeadNew South WalesAustralia
- School of Public Health, Faculty of Medicine and HealthUniversity of SydneyCamperdownNew South WalesAustralia
| | - Darren Lee
- Department of Renal MedicineEastern HealthBox HillVictoriaAustralia
- Eastern Health Clinical SchoolMonash UniversityClaytonVictoriaAustralia
- Department of NephrologyAustin HealthHeidelbergVictoriaAustralia
| | | | - Sanduni C. Fernando
- Department of NephrologyMonash Medical CentreClaytonVictoriaAustralia
- Centre for Inflammatory Diseases, Department of MedicineMonash UniversityClaytonVictoriaAustralia
| | - Stephanie Fang‐Tzu Kuo
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of MedicineThe University of MelbourneMelbourneVictoriaAustralia
| | - Sarah Tan
- Department of NephrologyFlinders Medical CentreAdelaideAustralia
| | - Sadia Jahan
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
| | - Wai H. Lim
- Department of Renal MedicineSir Charles Gairdner HospitalPerthAustralia
- Medical SchoolUniversity of Western AustraliaPerthAustralia
| |
Collapse
|
4
|
Malvica S, Fenaroli P, Lee CY, Louis S, Celia AI, Bagnasco S, Yang X, Hodgin JB, Buyon J, Magder L, Petri M, Rosenberg A, Fava A. Inflammation in Areas of Fibrosis Precedes Loss of Kidney Function in Lupus Nephritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625225. [PMID: 39651286 PMCID: PMC11623599 DOI: 10.1101/2024.11.25.625225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Interstitial fibrosis in lupus nephritis (LN) is often infiltrated by immune cells but typically regarded as nonspecific "scar reaction." This study aimed to investigate the relationship between inflammatory fibrosis and kidney disease progression in LN. Methods Interstitial fibrosis and tubular atrophy (IFTA) were scored in 124 LN kidney biopsies. Inflammation in areas of IFTA (i-IFTA) was graded 0-3 according to the Banff Classification of Allograft Pathology. Significant glomerular filtration rate (GFR) loss was defined as a decline of >15 ml/min at 3 years from biopsy. Immune cell phenotype was defined by serial immunohistochemistry (13-plex). Results IFTA was observed in 88/124 (71%) biopsies, and i-IFTA was identified in 76/88 (86%) cases. The distribution of i-IFTA grades was heterogenous across all IFTA grades. In patients with moderate-to-severe IFTA (>25%), the degree of i-IFTA was associated with a higher risk of significant GFR loss: 0/2 (0%), 1/3 (33%), 3/4 (75%), and 7/9 (78%) for i-IFTA grades 0, 1, 2, and 3, respectively (p = 0.028). Multiplexed histology revealed that i-IFTA was mostly composed of CD163+ macrophages and CD4 T cells, followed by CD8 T cells and granulocytes. Conclusion I-IFTA is frequently observed in LN and is dominated by macrophages and T cells. For patients with baseline IFTA >25%, the degree of i-IFTA emerged as a predictor of GFR loss. These data support the routine scoring of i-IFTA in LN due to its prognostic implications and nominate i-IFTA as a potential therapeutic target. LAY SUMMARY Scar tissue often contains immune cells, but we still do not fully understand their role. In lupus nephritis (LN), this is typically dismissed as "nonspecific inflammation". However, our study analyzed kidney biopsies from 124 people with LN and found that inflammation in scarred areas may predict future kidney function loss. Specifically, we identified a type of immune cell, CD163+ macrophages, that may contribute to scarring and kidney damage. Our findings suggest that routinely assessing inflammation in scarred areas could help predict kidney health in LN patients and highlight a possible new target for therapies to prevent kidney damage.
Collapse
|
5
|
Jonkman I, Jacobs MME, Negishi Y, Yanginlar C, Martens JHA, Baltissen M, Vermeulen M, van den Hoogen MWF, Baas M, van der Vlag J, Fayad ZA, Teunissen AJP, Madsen JC, Ochando J, Joosten LAB, Netea MG, Mulder WJM, Mhlanga MM, Hilbrands LB, Rother N, Duivenvoorden R. Trained immunity suppression determines kidney allograft survival. Am J Transplant 2024; 24:2022-2033. [PMID: 39147201 PMCID: PMC11789421 DOI: 10.1016/j.ajt.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
The innate immune system plays an essential role in regulating the immune responses to kidney transplantation, but the mechanisms through which innate immune cells influence long-term graft survival are unclear. The current study highlights the vital role of trained immunity in kidney allograft survival. Trained immunity describes the epigenetic and metabolic changes that innate immune cells undergo following an initial stimulus, allowing them have a stronger inflammatory response to subsequent stimuli. We stimulated healthy peripheral blood mononuclear cells with pretransplant and posttransplant serum of kidney transplant patients and immunosuppressive drugs in an in vitro trained immunity assay and measured tumor necrosis factor and interleukin 6 cytokine levels in the supernatant as a readout for trained immunity. We show that the serum of kidney transplant recipients collected 1 week after transplantation can suppress trained immunity. Importantly, we found that kidney transplant recipients whose serum most strongly suppressed trained immunity rarely experienced graft loss. This suppressive effect of posttransplant serum is likely mediated by previously unreported effects of immunosuppressive drugs. Our findings provide mechanistic insights into the role of innate immunity in kidney allograft survival, uncovering trained immunity as a potential therapeutic target for improving graft survival.
Collapse
Affiliation(s)
- Inge Jonkman
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maaike M E Jacobs
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yutaka Negishi
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands; Department of Cell Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Cansu Yanginlar
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Marijke Baltissen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Martijn W F van den Hoogen
- Department of Internal Medicine, Erasmus Medical Center Transplant Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marije Baas
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zahi A Fayad
- Department of Radiology, Biomolecular Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Abraham J P Teunissen
- Department of Radiology, Biomolecular Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joren C Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA; Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Genetics, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Willem J M Mulder
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Biomedical Engineering and Institute for Complex Molecular Systems, Laboratory of Chemical Biology, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Musa M Mhlanga
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands; Department of Cell Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Radiology, Biomolecular Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
6
|
Kalaria AL, Yamada T, Klein-Fedyshin M, Obata S, Cruz-Peralta M, Parrish B, Rahman AZ, Molinari M, Mehta RB. Subclinical rejection and allograft survival in kidney transplantation: protocol for a systematic review and meta-analysis. BMJ Open 2024; 14:e085098. [PMID: 39025816 PMCID: PMC11261677 DOI: 10.1136/bmjopen-2024-085098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
INTRODUCTION Subclinical rejection (SCR) refers to the presence of acute rejection without accompanying kidney allograft dysfunction. The impact of SCR on long-term graft survival remains a subject of ongoing debate. METHODS AND ANALYSIS We will perform a systematic search of databases including MEDLINE, Embase and Cochrane Central, from January 1995 to November 2023. We will include English-language studies involving adult kidney transplant patients who investigated SCR. We will exclude studies focused on 'for-cause' biopsies. Both title, abstract screening and full-text screening will be performed by two or more reviewers. The primary outcome of this study will be death-censored allograft loss. The secondary outcome will include development of subsequent rejection. For time-dependent outcomes, we will prioritise HRs and the 95% CIs. In cases where HRs are unavailable, we will calculate risk ratios based on the recorded events. The risk of bias will be assessed using the Cochrane Collaboration's revised tool for assessing the risk of bias in randomised trials and the Newcastle-Ottawa scale for cohort studies. We will employ a random effects model. We will evaluate heterogeneity using the I2 variable. We will assess publication bias by funnel plots, Begg and Mazumdar test, and Egger's test. ETHICS AND DISSEMINATION Ethics approval does not apply as no original data will be collected. The results will be disseminated through peer-reviewed publications and conference presentations. PROSPERO REGISTRATION NUMBER CRD42023463536.
Collapse
Affiliation(s)
- Arjun Lalit Kalaria
- Division of Transplant Nephrology, Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Takayuki Yamada
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, Washington, USA
| | | | - Shota Obata
- Department of Medicine, Mount Sinai Beth Israel, Icahn School of Medicine, New York, New York, USA
| | - Massiel Cruz-Peralta
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Bryce Parrish
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Amaan Z Rahman
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michele Molinari
- Department of Surgery, Division of Transplantation, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Rajil B Mehta
- Division of Transplant Nephrology, Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Mengel M, Adam BA. Emerging phenotypes in kidney transplant rejection. Curr Opin Organ Transplant 2024; 29:97-103. [PMID: 38032262 DOI: 10.1097/mot.0000000000001130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
PURPOSE OF REVIEW This review focuses on more recently emerging rejection phenotypes in the context of time post transplantation and the resulting differential diagnostic challenges. It also discusses how novel ancillary diagnostic tools can potentially increase the accuracy of biopsy-based rejection diagnosis. RECENT FINDINGS With advances in reducing immunological risk at transplantation and improved immunosuppression treatment renal allograft survival improved. However, allograft rejection remains a major challenge and represent a frequent course for allograft failure. With prolonged allograft survival, novel phenotypes of rejection are emerging, which can show complex overlap and transition between cellular and antibody-mediated rejection mechanisms as well as mixtures of acute/active and chronic diseases. With the emerging complexity in rejection phenotypes, it is crucial to achieve diagnostic accuracy in the individual patient. SUMMARY The prospective validation and adoption of novel molecular and computational diagnostic tools into well defined and appropriate clinical context of uses will improve our ability to accurately diagnose, stage, and grade allograft rejection.
Collapse
Affiliation(s)
- Michael Mengel
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
8
|
Alexander MP, Zaidi M, Larson N, Mullan A, Pavelko KD, Stegall MD, Bentall A, Wouters BG, McKee T, Taner T. Exploring the single-cell immune landscape of kidney allograft inflammation using imaging mass cytometry. Am J Transplant 2024; 24:549-563. [PMID: 37979921 DOI: 10.1016/j.ajt.2023.11.008] [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: 07/26/2023] [Revised: 11/01/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
Kidney allograft inflammation, mostly attributed to rejection and infection, is an important cause of graft injury and loss. Standard histopathological assessment of allograft inflammation provides limited insights into biological processes and the immune landscape. Here, using imaging mass cytometry with a panel of 28 validated biomarkers, we explored the single-cell landscape of kidney allograft inflammation in 32 kidney transplant biopsies and 247 high-dimensional histopathology images of various phenotypes of allograft inflammation (antibody-mediated rejection, T cell-mediated rejection, BK nephropathy, and chronic pyelonephritis). Using novel analytical tools, for cell segmentation, we segmented over 900 000 cells and developed a tissue-based classifier using over 3000 manually annotated kidney microstructures (glomeruli, tubules, interstitium, and arteries). Using PhenoGraph, we identified 11 immune and 9 nonimmune clusters and found a high prevalence of memory T cell and macrophage-enriched immune populations across phenotypes. Additionally, we trained a machine learning classifier to identify spatial biomarkers that could discriminate between the different allograft inflammatory phenotypes. Further validation of imaging mass cytometry in larger cohorts and with more biomarkers will likely help interrogate kidney allograft inflammation in more depth than has been possible to date.
Collapse
Affiliation(s)
- Mariam P Alexander
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| | - Mark Zaidi
- Department of Medical Biophysics, University of Toronto, Canada
| | - Nicholas Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Aidan Mullan
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin D Pavelko
- Immune Monitoring Core Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark D Stegall
- Departments of Surgery and Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew Bentall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Bradly G Wouters
- Department of Medical Biophysics, University of Toronto, Canada; Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Trevor McKee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Pathomics Inc., Toronto, Ontario, Canada
| | - Timucin Taner
- Departments of Surgery and Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Cristoferi I, Varol H, van Baardwijk M, Rahiem L, Lila KA, van den Bosch TPP, Baan CC, Hesselink DA, Kramann R, Minnee RC, Mustafa DAM, Reinders MEJ, Roelen DL, Shahzad-Arshad SP, Smith RN, Stubbs AP, Colvin RB, Rosales IA, Clahsen-van Groningen MC. Multiomic profiling of transplant glomerulopathy reveals a novel T-cell dominant subclass. Kidney Int 2024; 105:812-823. [PMID: 38128610 DOI: 10.1016/j.kint.2023.11.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/04/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Kidney transplant (KTx) biopsies showing transplant glomerulopathy (TG) (glomerular basement membrane double contours (cg) > 0) and microvascular inflammation (MVI) in the absence of C4d staining and donor-specific antibodies (DSAs) do not fulfill the criteria for chronic active antibody-mediated rejection (CA-AMR) diagnosis and do not fit into any other Banff category. To investigate this, we initiated a multicenter intercontinental study encompassing 36 cases, comparing the immunomic and transcriptomic profiles of 14 KTx biopsies classified as cg+MVI DSA-/C4d- with 22 classified as CA-AMR DSA+/C4d+ through novel transcriptomic analysis using the NanoString Banff-Human Organ Transplant (B-HOT) panel and subsequent orthogonal subset analysis using two innovative 5-marker multiplex immunofluorescent panels. Nineteen genes were differentially expressed between the two study groups. Samples diagnosed with CA-AMR DSA+/C4d+ showed a higher glomerular abundance of natural killer cells and higher transcriptomic cell type scores for macrophages in an environment characterized by increased expression of complement-related genes (i.e., C5AR1) and higher activity of angiogenesis, interstitial fibrosis tubular atrophy, CA-AMR, and DSA-related pathways when compared to samples diagnosed with cg+MVI DSA-/C4d-. Samples diagnosed with cg+MVI DSA-/C4d- displayed a higher glomerular abundance and activity of T cells (CD3+, CD3+CD8+, and CD3+CD8-). Thus, we show that using novel multiomic techniques, KTx biopsies with cg+MVI DSA-/C4d- have a prominent T-cell presence and activity, putting forward the possibility that these represent a more T-cell dominant phenotype.
Collapse
Affiliation(s)
- Iacopo Cristoferi
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands; Department of Surgery, Division of HPB & Transplant Surgery, Erasmus MC Transplant Institute, Rotterdam, the Netherlands.
| | - Hilal Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Myrthe van Baardwijk
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands; Department of Surgery, Division of HPB & Transplant Surgery, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Layla Rahiem
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Karishma A Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Rafael Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC Transplant Institute, Rotterdam, the Netherlands; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Department of Nephrology and Clinical Immunology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Robert C Minnee
- Department of Surgery, Division of HPB & Transplant Surgery, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology and Clinical Bioinformatics, the Tumor Immuno-Pathology Laboratory, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Shazia P Shahzad-Arshad
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Rex N Smith
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew P Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands
| | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marian C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Transplant Institute, Rotterdam, the Netherlands; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
10
|
Peng L, Wang C, Yu S, Li Q, Wu G, Lai W, Min J, Chen G. Dysregulated lipid metabolism is associated with kidney allograft fibrosis. Lipids Health Dis 2024; 23:37. [PMID: 38308271 PMCID: PMC10837934 DOI: 10.1186/s12944-024-02021-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/17/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Interstitial fibrosis and tubular atrophy (IF/TA), a histologic feature of kidney allograft destruction, is linked to decreased allograft survival. The role of lipid metabolism is well-acknowledged in the area of chronic kidney diseases; however, its role in kidney allograft fibrosis is still unclarified. In this study, how lipid metabolism contributes to kidney allografts fibrosis was examined. METHODS A comprehensive bioinformatic comparison between IF/TA and normal kidney allograft in the Gene Expression Omnibus (GEO) database was conducted. Further validations through transcriptome profiling or pathological staining of human recipient biopsy samples and in rat models of kidney transplantation were performed. Additionally, the effects of enhanced lipid metabolism on changes in the fibrotic phenotype induced by TGF-β1 were examined in HK-2 cell. RESULTS In-depth analysis of the GEO dataset revealed a notable downregulation of lipid metabolism pathways in human kidney allografts with IF/TA. This decrease was associated with increased level of allograft rejection, inflammatory responses, and epithelial mesenchymal transition (EMT). Pathway enrichment analysis showed the downregulation in mitochondrial LC-fatty acid beta-oxidation, fatty acid beta-oxidation (FAO), and fatty acid biosynthesis. Dysregulated fatty acid metabolism was also observed in biopsy samples from human kidney transplants and in fibrotic rat kidney allografts. Notably, the areas affected by IF/TA had increased immune cell infiltration, during which increased EMT biomarkers and reduced CPT1A expression, a key FAO enzyme, were shown by immunohistochemistry. Moreover, under TGF-β1 induction, activating CPT1A with the compound C75 effectively inhibited migration and EMT process in HK-2 cells. CONCLUSIONS This study reveal a critical correlation between dysregulated lipid metabolism and kidney allograft fibrosis. Enhancing lipid metabolism with CPT1A agonists could be a therapeutic approach to mitigate kidney allografts fibrosis.
Collapse
Affiliation(s)
- Linjie Peng
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Chang Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Shuangjin Yu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Qihao Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Guobin Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Weijie Lai
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Jianliang Min
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China
| | - Guodong Chen
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- The First Affiliated Hospital, Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
Landsberg A, Raza SS, Seifert ME, Blydt-Hansen TD. Follow-up biopsies identify high rates of persistent rejection in pediatric kidney transplant recipients after treatment of T cell-mediated rejection. Pediatr Transplant 2024; 28:e14617. [PMID: 37750353 PMCID: PMC10872996 DOI: 10.1111/petr.14617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Incomplete resolution of T cell-mediated rejection (TCMR) after treatment may not be detected with serum creatinine monitoring and is associated with donor-specific antibodies and chronic rejection. We evaluate the utility of follow-up biopsies (FUB) to identify and characterize rates of persistent TCMR after treatment in pediatric kidney transplant patients. METHODS Patients from two pediatric transplant centers performing standard of care FUB at 1.5-2 months after treatment for TCMR were included. FUB were evaluated for extent of rejection resolution (complete vs. incomplete) and grade. Clinical data at time of FUB and later were reported, where available. RESULTS Fifty-eight patients underwent FUB, at mean of 1.7 months (SD 0.7) post-index biopsy. Rejection grade on index biopsy was Banff borderline (≥i1t1 and CONCLUSIONS FUB were effective at detecting persistent rejection, which was common among pediatric transplant patients after standard TCMR treatment. Until more effective rejection treatments or sensitive biomarkers are available, FUB may be effectively utilized to identify patients with ongoing rejection who would benefit from further treatment.
Collapse
Affiliation(s)
- Adina Landsberg
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - S. Sikandar Raza
- Department of Pediatrics, University of Alabama Heersink School of Medicine, Birmingham, AL, United States
| | - Michael E. Seifert
- Department of Pediatrics, University of Alabama Heersink School of Medicine, Birmingham, AL, United States
| | - Tom D. Blydt-Hansen
- BC Children’s Hospital, Division of Nephrology, Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Heldal TF, Åsberg A, Ueland T, Reisæter AV, Pischke SE, Mollnes TE, Aukrust P, Reinholt F, Hartmann A, Heldal K, Jenssen TG. Systemic inflammation early after kidney transplantation is associated with long-term graft loss: a cohort study. Front Immunol 2023; 14:1253991. [PMID: 37849758 PMCID: PMC10577420 DOI: 10.3389/fimmu.2023.1253991] [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: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
Background Early graft loss following kidney transplantation is mainly a result of acute rejection or surgical complications, while long-term kidney allograft loss is more complex. We examined the association between systemic inflammation early after kidney transplantation and long-term graft loss, as well as correlations between systemic inflammation scores and inflammatory findings in biopsies 6 weeks and 1 year after kidney transplantation. Methods We measured 21 inflammatory biomarkers 10 weeks after transplantation in 699 patients who were transplanted between 2009 and 2012 at Oslo University Hospital, Rikshospitalet, Norway. Low-grade inflammation was assessed with predefined inflammation scores based on specific biomarkers: one overall inflammation score and five pathway-specific scores. Surveillance or indication biopsies were performed in all patients 6 weeks after transplantation. The scores were tested in Cox regression models. Results Median follow-up time was 9.1 years (interquartile range 7.6-10.7 years). During the study period, there were 84 (12.2%) death-censored graft losses. The overall inflammation score was associated with long-term kidney graft loss both when assessed as a continuous variable (hazard ratio 1.03, 95% CI 1.01-1.06, P = 0.005) and as a categorical variable (4th quartile: hazard ratio 3.19, 95% CI 1.43-7.10, P = 0.005). In the pathway-specific analyses, fibrogenesis activity and vascular inflammation stood out. The vascular inflammation score was associated with inflammation in biopsies 6 weeks and 1 year after transplantation, while the fibrinogenesis score was associated with interstitial fibrosis and tubular atrophy. Conclusion In conclusion, a systemic inflammatory environment early after kidney transplantation was associated with biopsy-confirmed kidney graft pathology and long-term kidney graft loss. The systemic vascular inflammation score correlated with inflammatory findings in biopsies 6 weeks and 1 year after transplantation.
Collapse
Affiliation(s)
- Torbjørn F. Heldal
- Department of Internal Medicine, Telemark Hospital Trust, Skien, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
- Research Institute of Internal Medicine, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Anna V. Reisæter
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Søren E. Pischke
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Anesthesiology, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Tom E. Mollnes
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
- Research Institute of Internal Medicine, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Finn Reinholt
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anders Hartmann
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Kristian Heldal
- Department of Internal Medicine, Telemark Hospital Trust, Skien, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Trond G. Jenssen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| |
Collapse
|
13
|
Mubarak M, Raza A, Rashid R, Shakeel S. Evolution of human kidney allograft pathology diagnostics through 30 years of the Banff classification process. World J Transplant 2023; 13:221-238. [PMID: 37746037 PMCID: PMC10514746 DOI: 10.5500/wjt.v13.i5.221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 09/15/2023] Open
Abstract
The second half of the previous century witnessed a tremendous rise in the number of clinical kidney transplants worldwide. This activity was, however, accompanied by many issues and challenges. An accurate diagnosis and appropriate management of causes of graft dysfunction were and still are, a big challenge. Kidney allograft biopsy played a vital role in addressing the above challenge. However, its interpretation was not standardized for many years until, in 1991, the Banff process was started to fill this void. Thereafter, regular Banff meetings took place every 2 years for the past 30 years. Marked changes have taken place in the interpretation of kidney allograft biopsies, diagnosis, and classification of rejection and other non-rejection pathologies from the original Banff 93 classification. This review attempts to summarize those changes for increasing the awareness and understanding of kidney allograft pathology through the eyes of the Banff process. It will interest the transplant surgeons, physicians, pathologists, and allied professionals associated with the care of kidney transplant patients.
Collapse
Affiliation(s)
- Muhammed Mubarak
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Amber Raza
- Department of Nephrology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Rahma Rashid
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Shaheera Shakeel
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| |
Collapse
|
14
|
Masutani K. Progress in Pathological Diagnosis after Kidney Transplantation: Current Trend and Future Perspective. J Atheroscler Thromb 2023; 30:720-732. [PMID: 37245995 PMCID: PMC10322740 DOI: 10.5551/jat.rv22005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 05/30/2023] Open
Abstract
Advances in immunosuppressive therapy; posttransplant management of allograft rejection; and measures against infectious diseases, cardiovascular diseases, and malignancy dramatically improved graft and patient survival after kidney transplantation (KT). Among them, kidney allograft biopsy is an important tool and the gold standard for the diagnosis of various kidney allograft injuries, including allograft rejection, virus-induced nephropathy, calcineurin inhibitor toxicity, and posttransplant glomerular diseases. The Banff Conference on Allograft Pathology has contributed to establishing the diagnostic criteria for kidney allograft rejection and polyomavirus-associated nephropathy that are used as a common standard worldwide. In addition to the for-cause biopsy, many transplant centers perform protocol biopsies in the early and late posttransplant periods to detect and treat allograft injury earlier. Preimplantation biopsy in deceased-donor KT has also been performed, especially in the marginal donor, and attempts have been made to predict the prognosis in combination with clinical information and the renal resistance of hypothermic machine perfusion. Regarding the preimplantation biopsy from a living kidney donor, it can provide useful information on aging and/or early changes in lifestyle diseases, such as glomerulosclerosis, tubulointerstitial changes, and arterial and arteriolar sclerosis, and be used as a reference for the subsequent management of living donors. In this review, morphologic features of important kidney allograft pathology, such as allograft rejection and polyomavirus-associated nephropathy, according to the latest Banff classification and additional information derived from protocol biopsy, and future perspectives with recently developed technologies are discussed.
Collapse
Affiliation(s)
- Kosuke Masutani
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka,
Japan
| |
Collapse
|
15
|
Yadav B, Prasad N, Agrawal V, Agarwal V, Jain M. Lower Circulating Cytotoxic T-Cell Frequency and Higher Intragraft Granzyme-B Expression Are Associated with Inflammatory Interstitial Fibrosis and Tubular Atrophy in Renal Allograft Recipients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1175. [PMID: 37374379 DOI: 10.3390/medicina59061175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: Inflammatory interstitial fibrosis and tubular atrophy (i-IFTA) is an inflammation in the area of tubular atrophy and fibrosis. i-IFTA is poorly associated with graft outcome and associated with infiltration of inflammatory mononuclear cells. A cytotoxic T cell is a granzyme B+CD8+CD3+ T cell, mainly secret granzyme B. Granzyme B is a serine protease that may mediate allograft injury and inflammatory interstitial fibrosis and tubular atrophy (i-IFTA). However, there is no report identifying the association of granzyme B with i-IFTA after a long post-transplant interval. Material and Methods: In this study, we have measured the cytotoxic T-cell frequency with flow cytometry, serum and PBMCs culture supernatants granzyme-B levels with ELISA and intragraft granzyme-B mRNA transcript expression with the RT-PCR in RTRs in 30 patients with biopsy-proven i-IFTA and 10 patients with stable graft function. Result: The frequency of cytotoxic T cells (CD3+CD8+ granzyme B+) in SGF vs. i-IFTA was (27.96 ± 4.86 vs. 23.19 ± 3.85%, p = 0.011), the serum granzyme-B level was (100.82 ± 22.41 vs. 130.32 ± 46.60, p = 0.038 pg/mL) and the intragraft granzyme-B mRNA transcript expression was (1.01 ± 0.048 vs. 2.10 ± 1.02, p < 0.001 fold). The frequency of CD3+ T cells in SGF vs. i-IFTA was (66.08 ± 6.8 vs. 65.18 ± 9.35%; p = 0.68) and that of CD3+CD8+ T cells was (37.29 ± 4.11 vs. 34.68 ± 5.43%; p = 0.28), which were similar between the 2 groups. CTLc frequency was negatively correlated with urine proteinuria (r = -0.51, p < 0.001), serum creatinine (r = -0.28, p = 0.007) and eGFR (r = -0.28, p = 0.037). Similarly, the PBMC culture supernatants granzyme-B level was negatively correlated with urine proteinuria (r = -0.37, p < 0.001) and serum creatinine (r = -0.31, p = 0.002), while the serum granzyme-B level (r = 0.343, p = 0.001) and intragraft granzyme-B mRNA transcript expression (r = 0.38, p < 0.001) were positively correlated with proteinuria. Conclusions: A decrease in the CTLc frequency in circulation and an increased serum granzyme-B level and intragraft granzyme-B mRNA expression shows that cytotoxic T cells may mediate the allograft injury in RTRs with i-IFTA by releasing granzyme B in serum and intragraft tissue.
Collapse
Affiliation(s)
- Brijesh Yadav
- Department of Nephrology and Renal Transplantation, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Narayan Prasad
- Department of Nephrology and Renal Transplantation, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Vinita Agrawal
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Vikas Agarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Manoj Jain
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
16
|
Yamamoto I, Kawabe M, Hayashi A, Kobayashi A, Yamamoto H, Yokoo T. Challenges Posed by the Banff Classification: Diagnosis and Treatment of Chronic Active T-Cell-Mediated Rejection. Nephron Clin Pract 2023; 147 Suppl 1:74-79. [PMID: 36928337 DOI: 10.1159/000530158] [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: 12/15/2022] [Accepted: 02/14/2023] [Indexed: 03/18/2023] Open
Abstract
The three primary sites of acute T-cell-mediated rejection (TCMR) in transplanted kidneys are the tubular epithelial cells, interstitium, and the vascular endothelial cells. The pathology of acute lesions is characterized by inflammatory cell infiltration; the final diagnosis suggested by the Banff 2019 classification is guided by grading of tubulitis (the t score), interstitial inflammation (the i score), and endarteritis (the v score). Consistent major issues when using the Banff classification are the etiological classifications of interstitial fibrosis and tubular atrophy (IFTA). From 2015 to 2019, technological advances (i.e., genetic analysis in paraffin sections) increased our understanding of IFTA status in patients with smoldering acute TCMR and the roles played by inflammatory cell infiltration (the i-IFTA score) and tubulitis (the t-IFTA score) in IFTA. These two scores were introduced when establishing the diagnostic criteria for chronic active TCMR. Despite the increase in complexity and the lack of a consensus treatment for chronic active TCMR, the Banff classification may evolve as new techniques (i.e., genetic analysis in paraffin sections and deep learning of renal pathology) are introduced. The Banff conference proceeded as follows. First, lesions were defined. Next, working groups were established to better understand the lesions and to derive better classification methods. Finally, the new Banff classification was developed. This approach will continue to evolve; the Banff classification will become a very useful diagnostic standard. This paper overviews the history of TCMR diagnosis using the Banff classification, and the clinical importance, treatment, and prospects for acute and chronic active TCMR.
Collapse
Affiliation(s)
- Izumi Yamamoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Mayuko Kawabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayaka Hayashi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Akimitsu Kobayashi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyasu Yamamoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
van der Elst G, Varol H, Hermans M, Baan CC, Duong-van Huyen JP, Hesselink DA, Kramann R, Rabant M, Reinders MEJ, von der Thüsen JH, van den Bosch TPP, Clahsen-van Groningen MC. The mast cell: A Janus in kidney transplants. Front Immunol 2023; 14:1122409. [PMID: 36891297 PMCID: PMC9986315 DOI: 10.3389/fimmu.2023.1122409] [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: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Mast cells (MCs) are innate immune cells with a versatile set of functionalities, enabling them to orchestrate immune responses in various ways. Aside from their known role in allergy, they also partake in both allograft tolerance and rejection through interaction with regulatory T cells, effector T cells, B cells and degranulation of cytokines and other mediators. MC mediators have both pro- and anti-inflammatory actions, but overall lean towards pro-fibrotic pathways. Paradoxically, they are also seen as having potential protective effects in tissue remodeling post-injury. This manuscript elaborates on current knowledge of the functional diversity of mast cells in kidney transplants, combining theory and practice into a MC model stipulating both protective and harmful capabilities in the kidney transplant setting.
Collapse
Affiliation(s)
- G van der Elst
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - H Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M Hermans
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - C C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - D A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - R Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - M Rabant
- Department of Pathology, Necker Hospital, APHP, Paris, France
| | - M E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
18
|
The Histological Spectrum and Clinical Significance of T Cell-mediated Rejection of Kidney Allografts. Transplantation 2022; 107:1042-1055. [PMID: 36584369 DOI: 10.1097/tp.0000000000004438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T cell-mediated rejection (TCMR) remains a significant cause of long-term kidney allograft loss, either indirectly through induction of donor-specific anti-HLA alloantibodies or directly through chronic active TCMR. Whether found by indication or protocol biopsy, Banff defined acute TCMR should be treated with antirejection therapy and maximized maintenance immunosuppression. Neither isolated interstitial inflammation in the absence of tubulitis nor isolated tubulitis in the absence of interstitial inflammation results in adverse outcomes, and neither requires antirejection treatment. RNA gene expression analysis of biopsy material may supplement conventional histology, especially in ambiguous cases. Lesser degrees of tubular and interstitial inflammation (Banff borderline) may portend adverse outcomes and should be treated when found on an indication biopsy. Borderline lesions on protocol biopsies may resolve spontaneously but require close follow-up if untreated. Following antirejection therapy of acute TCMR, surveillance protocol biopsies should be considered. Minimally invasive blood-borne assays (donor-derived cell-free DNA and gene expression profiling) are being increasingly studied as a means of following stable patients in lieu of biopsy. The clinical benefit and cost-effectiveness require confirmation in randomized controlled trials. Treatment of acute TCMR is not standardized but involves bolus corticosteroids with lymphocyte depleting antibodies for severe, refractory, or relapsing cases. Arteritis may be found with acute TCMR, active antibody-mediated rejection, or mixed rejections and should be treated accordingly. The optimal treatment ofchronic active TCMR is uncertain. Randomized controlled trials are necessary to optimally define therapy.
Collapse
|
19
|
Mehta RB, Melgarejo I, Viswanathan V, Zhang X, Pittappilly M, Randhawa P, Puttarajappa C, Sood P, Wu C, Sharma A, Molinari M, Hariharan S. Long-term immunological outcomes of early subclinical inflammation on surveillance kidney allograft biopsies. Kidney Int 2022; 102:1371-1381. [PMID: 36049641 DOI: 10.1016/j.kint.2022.07.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 01/12/2023]
Abstract
The long-term impact of early subclinical inflammation (SCI) through surveillance biopsy has not been well studied. To do this, we recruited a prospective observational cohort that included 1000 sequential patients who received a kidney transplant from 2013-2017 at our center. A total of 586 patients who underwent a surveillance biopsy in their first year post-transplant were included after excluding those with clinical rejections, and those who were unable to undergo a surveillance biopsy. Patients were classified based on their biopsy findings: 282 with NSI (No Significant Inflammation) and 304 with SCI-T (SCI and Tubulitis) which was further subdivided into 182 with SC-BLR (Subclinical Borderline Changes) and 122 with SC-TCMR (Subclinical T Cell Mediated Rejection, Banff 2019 classification of 1A or more). We followed the clinical and immunological events including Clinical Biopsy Proven Acute Rejection [C-BPAR], long-term kidney function and death-censored graft loss over a median follow-up of five years. Episodes of C-BPAR were noted at a median of two years post-transplant. Adjusted odds of having a subsequent C-BPAR was significantly higher in the SCI-T group [SC-BLR and SC-TCMR] compared to NSI 3.8 (2.1-7.5). The adjusted hazard for death-censored graft loss was significantly higher with SCI-T compared to NSI [1.99 (1.04-3.84)]. Overall, SCI detected through surveillance biopsy within the first year post-transplant is a harbinger for subsequent immunological events and is associated with a significantly greater hazard for subsequent C-BPAR and death-censored graft loss. Thus, our study highlights the need for identifying patients with SCI through surveillance biopsy and develop strategies to prevent further alloimmune injuries.
Collapse
Affiliation(s)
- Rajil B Mehta
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Ivy Melgarejo
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Vignesh Viswanathan
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Xingyu Zhang
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Matthew Pittappilly
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Parmjeet Randhawa
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Chethan Puttarajappa
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Puneet Sood
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Christine Wu
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Akhil Sharma
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michele Molinari
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sundaram Hariharan
- Division of Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Renal Fibrosis in Lupus Nephritis. Int J Mol Sci 2022; 23:ijms232214317. [PMID: 36430794 PMCID: PMC9699516 DOI: 10.3390/ijms232214317] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Fibrosis can be defined as a pathological process in which deposition of connective tissue replaces normal parenchyma. The kidney, like any organ or tissue, can be impacted by this maladaptive reaction, resulting in persistent inflammation or long-lasting injury. While glomerular injury has traditionally been regarded as the primary focus for classification and prognosis of lupus nephritis (LN), increasing attention has been placed on interstitial fibrosis and tubular atrophy as markers of injury severity, predictors of therapeutic response, and prognostic factors of renal outcome in recent years. This review will discuss the fibrogenesis in LN and known mechanisms of renal fibrosis. The importance of the chronicity index, which was recently added to the histological categorization of LN, and its role in predicting treatment response and renal prognosis for patients with LN, will be explored. A better understanding of cellular and molecular pathways involved in fibrosis in LN could enable the identification of individuals at higher risk of progression to chronic kidney disease and end-stage renal disease, and the development of new therapeutic strategies for lupus patients.
Collapse
|
21
|
Guy P, Delas A, Esposito L, Cointault O, Colombat M, Congy-Jolivet N, Raynaud M, Kamar N, Del Bello A. Progression of histological lesions after ABO incompatible kidney transplantation. Front Immunol 2022; 13:969998. [PMID: 36275771 PMCID: PMC9582152 DOI: 10.3389/fimmu.2022.969998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022] Open
Abstract
Recent large meta-analyses suggested a poorer long-term patients’ and grafts’ outcomes after ABO incompatible (ABOi) living-donor kidney transplantation (LDKT) compared to ABO compatible LDKT. However, little is known about the long-term histological pattern after ABOi LDKT. We compared the histological features observed on protocol biopsies from 03/11 to 11/19 in 94 ABOi LDKT (including 14 with preformed Donor Specific Antibodies, pDSAs), 27 LDKT ABO compatible (ABOc) with pDSAs, and 21 ABOc without pDSAs) during the first five years post transplantation. During the first 5 years post-transplantation, a progression of chronic lesions (patients with a ci >0 raised from 11% to 65%, p<0.0001, patients with a ct >0 raised from 29% to 78%, p<0.0001) was observed in ABOi LDKT without pDSAs. Histological patterns of evolution were comparable to those observed in ABOc kidney transplant patients. Microvascular inflammation was lower in ABOi LDKT without pDSAs compared to those with pDSAs (ABOi or ABOc). At last follow-up, 28 months, IQR (15-48) post-transplantation, 29 patients (36%) had a severe graft dysfunction (defined by a CKD-epi eGFR < 30 mL/min/1.73m²). The donor age was a predictive factor for the development of severe kidney allograft dysfunction at last follow-up (HR= 1.05, 95% CI [1.05-1.10], p= 0.03). Hence, long-term histological analysis of ABOi LDKT shows only an increase of chronic interstitial and tubular atrophy changes, without active lesions. These data confirm that ABOi LDKT programs can be securely developed.
Collapse
Affiliation(s)
- Pierre Guy
- Department of Nephrology and Organ Transplantation, Centre Hospitalier et Universitaire (CHU), Toulouse, France
| | - Audrey Delas
- Department of Pathology, Toulouse University Hospital, Toulouse, France
| | - Laure Esposito
- Department of Nephrology and Organ Transplantation, Centre Hospitalier et Universitaire (CHU), Toulouse, France
| | - Olivier Cointault
- Department of Nephrology and Organ Transplantation, Centre Hospitalier et Universitaire (CHU), Toulouse, France
| | - Magali Colombat
- Department of Pathology, Toulouse University Hospital, Toulouse, France
- Centre Hospitalier et Universitaire, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Nicolas Congy-Jolivet
- Laboratory of Immunology, Biology Department, Centre Hospitalier et Universitaire (CHU) de Toulouse, Toulouse, France
| | - Marc Raynaud
- Paris Translational Research Epidemiology and Biostatistics Department, Université de Paris, INSERM U970, PARCC, Paris, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, Centre Hospitalier et Universitaire (CHU), Toulouse, France
- Centre Hospitalier et Universitaire, Université Paul Sabatier Toulouse III, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1043-CNRS 5282, Toulouse, France
| | - Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, Centre Hospitalier et Universitaire (CHU), Toulouse, France
- Centre Hospitalier et Universitaire, Université Paul Sabatier Toulouse III, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1043-CNRS 5282, Toulouse, France
- *Correspondence: Arnaud Del Bello,
| |
Collapse
|
22
|
Hermsen M, Ciompi F, Adefidipe A, Denic A, Dendooven A, Smith BH, van Midden D, Bräsen JH, Kers J, Stegall MD, Bándi P, Nguyen T, Swiderska-Chadaj Z, Smeets B, Hilbrands LB, van der Laak JAWM. Convolutional Neural Networks for the Evaluation of Chronic and Inflammatory Lesions in Kidney Transplant Biopsies. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1418-1432. [PMID: 35843265 DOI: 10.1016/j.ajpath.2022.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 06/13/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
In kidney transplant biopsies, both inflammation and chronic changes are important features that predict long-term graft survival. Quantitative scoring of these features is important for transplant diagnostics and kidney research. However, visual scoring is poorly reproducible and labor intensive. The goal of this study was to investigate the potential of convolutional neural networks (CNNs) to quantify inflammation and chronic features in kidney transplant biopsies. A structure segmentation CNN and a lymphocyte detection CNN were applied on 125 whole-slide image pairs of periodic acid-Schiff- and CD3-stained slides. The CNN results were used to quantify healthy and sclerotic glomeruli, interstitial fibrosis, tubular atrophy, and inflammation within both nonatrophic and atrophic tubuli, and in areas of interstitial fibrosis. The computed tissue features showed high correlation with Banff lesion scores of five pathologists (A.A., A.Dend., J.H.B., J.K., and T.N.). Analyses on a small subset showed a moderate correlation toward higher CD3+ cell density within scarred regions and higher CD3+ cell count inside atrophic tubuli correlated with long-term change of estimated glomerular filtration rate. The presented CNNs are valid tools to yield objective quantitative information on glomeruli number, fibrotic tissue, and inflammation within scarred and non-scarred kidney parenchyma in a reproducible manner. CNNs have the potential to improve kidney transplant diagnostics and will benefit the community as a novel method to generate surrogate end points for large-scale clinical studies.
Collapse
Affiliation(s)
- Meyke Hermsen
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Adeyemi Adefidipe
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Aleksandar Denic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Amélie Dendooven
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Faculty of Medicine, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Byron H Smith
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota; Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Dominique van Midden
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Jesper Kers
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands; Center for Analytical Sciences Amsterdam, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Mark D Stegall
- Division of Transplantation Surgery, Mayo Clinic, Rochester, Minnesota
| | - Péter Bándi
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tri Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Zaneta Swiderska-Chadaj
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands; Faculty of Electrical Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Bart Smeets
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen A W M van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands; Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden.
| |
Collapse
|
23
|
Etta DPK. Implications of Banff Classification Schema: A Journey of Three Decades. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2022; 33:598-602. [PMID: 37929555 DOI: 10.4103/1319-2442.388196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Affiliation(s)
- Dr Praveen Kumar Etta
- Department of Nephrology and Renal Transplantation, TX Hospitals, Hyderabad, Telangana, India., India
| |
Collapse
|
24
|
Schuster A, Steines L, Müller K, Zeman F, Findeisen P, Banas B, Bergler T. Dickkopf 3-A New Indicator for the Deterioration of Allograft Function After Kidney Transplantation. Front Med (Lausanne) 2022; 9:885018. [PMID: 35646976 PMCID: PMC9130628 DOI: 10.3389/fmed.2022.885018] [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: 02/27/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Evidence of tubular atrophy and interstitial fibrosis is prognostically unfavorable and associated with a premature graft loss after kidney transplantation. Recently, Dickkopf 3 (DKK3), a profibrotic glycoprotein released by stressed tubular epithelial cells, has been identified to cause IF/TA by regulating the Wnt/β-catenin signaling and seems to engage a T-cell response. The aim of our study was to determine if a correlation between DKK3 and graft function exists and if DKK3 could be a new indicator to identify patients at risk for a deterioration in graft function. Patients, transplanted between 2016 and 2018, were analyzed with regard to DKK3 in the urine and graft function (creatinine, eGFR, albuminuria). Multivariable analyzes were used including known factors influencing graft function (PRA, donor age) to stress robustness of DKK3. The 3 and 12 month DKK3 values were significant predictors for subsequent graft function up to 36 months. An increase of DKK3 from month 3 to 12 of ≥ 25% showed a higher risk of an impaired graft function, with, e.g., a reduction in eGFR of about 9–10 ml/min in contrast to patients without intensified DKK3 increase. Induction therapy has an influence on DKK3 as patients induced with a T-cell depleting therapy showed a trend toward lower DKK3 values. In summary, our study is the first investigation of DKK3 in kidney transplant recipients and was able to show that DKK3 could forecast graft function. It is recommended to investigate the potential of DKK3 as a predictor of kidney function after transplantation in further studies.
Collapse
Affiliation(s)
- Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Karolina Müller
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | | | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
25
|
Becker JU, Seron D, Rabant M, Roufosse C, Naesens M. Evolution of the Definition of Rejection in Kidney Transplantation and Its Use as an Endpoint in Clinical Trials. Transpl Int 2022; 35:10141. [PMID: 35669978 PMCID: PMC9163319 DOI: 10.3389/ti.2022.10141] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
This article outlines the evolving definition of rejection following kidney transplantation. The viewpoints and evidence presented were included in documentation prepared for a Broad Scientific Advice request to the European Medicines Agency (EMA), relating to clinical trial endpoints in kidney transplantation. This request was initiated by the European Society for Organ Transplantation (ESOT) in 2016 and finalized following discussions between the EMA and ESOT in 2020. In ESOT’s opinion, the use of “biopsy-proven acute rejection” as an endpoint for clinical trials in kidney transplantation is no longer accurate, although it is still the approved histopathological endpoint. The spectrum of rejection is now divided into the phenotypes of borderline changes, T cell-mediated rejection, and antibody-mediated rejection, with the latter two phenotypes having further subclassifications. Rejection is also described in relation to graft (dys)function, diagnosed because of protocol (surveillance) or indication (for-cause) biopsies. The ongoing use of outdated terminology has become a potential barrier to clinical research in kidney transplantation. This article presents these perspectives and issues, and provides a foundation on which subsequent articles within this Special Issue of Transplant International build.
Collapse
Affiliation(s)
- Jan Ulrich Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Daniel Seron
- Department of Nephrology and Kidney Transplantation, Vall d’Hebrón University Hospital, Barcelona, Spain
| | - Marion Rabant
- Department of Pathology, Hôpital Necker–Enfants Malades, Paris, France
| | - Candice Roufosse
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| |
Collapse
|
26
|
De Lorenzo SB, Vrieze AM, Johnson RA, Lien KR, Nath KA, Garovic VD, Khazaie K, Grande JP. KLF11 deficiency enhances chemokine generation and fibrosis in murine unilateral ureteral obstruction. PLoS One 2022; 17:e0266454. [PMID: 35413089 PMCID: PMC9004740 DOI: 10.1371/journal.pone.0266454] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Progression of virtually all forms of chronic kidney disease (CKD) is associated with activation of pro-inflammatory and pro-fibrotic signaling pathways. Despite extensive research, progress in identifying therapeutic targets to arrest or slow progression of CKD has been limited by incomplete understanding of basic mechanisms underlying renal inflammation and fibrosis in CKD. Recent studies have identified Kruppel-like transcription factors that have been shown to play critical roles in renal development, homeostasis, and response to injury. Although KLF11 deficiency has been shown to increase collagen production in vitro and tissue fibrosis in other organs, no previous study has linked KLF11 to the development of CKD. We sought to test the hypothesis that KLF11 deficiency promotes CKD through upregulation of pro-inflammatory and pro-fibrogenic signaling pathways in murine unilateral ureteral obstruction (UUO), a well-established model of renal fibrosis. We found that KLF11-deficiency exacerbates renal injury in the UUO model through activation of the TGF-β/SMAD signaling pathway and through activation of several pro-inflammatory chemokine signaling pathways. Based on these considerations, we conclude that agents increase KLF11 expression may provide novel therapeutic targets to slow the progression of CKD.
Collapse
Affiliation(s)
- Silvana B. De Lorenzo
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alyssa M. Vrieze
- Department of Comparative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ruth A. Johnson
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karen R. Lien
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karl A. Nath
- Division of Nephrology & Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Vesna D. Garovic
- Division of Nephrology & Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Khashayarsha Khazaie
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Joseph P. Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ho J, Okoli GN, Rabbani R, Lam OLT, Reddy V, Askin N, Rampersad C, Trachtenberg A, Wiebe C, Nickerson P, Abou‐Setta AM. Effectiveness of T cell-mediated rejection therapy: A systematic review and meta-analysis. Am J Transplant 2022; 22:772-785. [PMID: 34860468 PMCID: PMC9300092 DOI: 10.1111/ajt.16907] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 01/25/2023]
Abstract
The effectiveness of T cell-mediated rejection (TCMR) therapy for achieving histological remission remains undefined in patients on modern immunosuppression. We systematically identified, critically appraised, and summarized the incidence and histological outcomes after TCMR treatment in patients on tacrolimus (Tac) and mycophenolic acid (MPA). English-language publications were searched in MEDLINE (Ovid), Embase (Ovid), Cochrane Central (Ovid), CINAHL (EBSCO), and Clinicaltrials.gov (NLM) up to January 2021. Study quality was assessed with the National Institutes of Health Study Quality Tool. We pooled results using an inverse variance, random-effects model and report the binomial proportions with associated 95% confidence intervals (95% CI). Statistical heterogeneity was explored using the I2 statistic. From 2875 screened citations, we included 12 studies (1255 participants). Fifty-eight percent were good/high quality while the rest were moderate quality. Thirty-nine percent of patients (95% CI 0.26-0.53, I2 77%) had persistent ≥Banff Borderline TCMR 2-9 months after anti-rejection therapy. Pulse steroids and augmented maintenance immunosuppression were mainstays of therapy, but considerable practice heterogeneity was present. A high proportion of biopsy-proven rejection exists after treatment emphasizing the importance of histology to characterize remission. Anti-rejection therapy is foundational to transplant management but well-designed clinical trials in patients on Tac/MPA immunosuppression are lacking to define the optimal therapeutic approach.
Collapse
Affiliation(s)
- Julie Ho
- Department of Internal MedicineMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - George N. Okoli
- George and Fay Yee Centre for Healthcare InnovationMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Rasheda Rabbani
- George and Fay Yee Centre for Healthcare InnovationMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada,Department of Community Health SciencesMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Otto L. T. Lam
- George and Fay Yee Centre for Healthcare InnovationMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Viraj K. Reddy
- George and Fay Yee Centre for Healthcare InnovationMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Nicole Askin
- Neil John Maclean Health Sciences LibraryUniversity of ManitobaWinnipegManitobaCanada
| | - Christie Rampersad
- Department of Internal MedicineMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Aaron Trachtenberg
- Department of Internal MedicineMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Chris Wiebe
- Department of Internal MedicineMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Peter Nickerson
- Department of Internal MedicineMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Ahmed M. Abou‐Setta
- George and Fay Yee Centre for Healthcare InnovationMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada,Department of Community Health SciencesMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
28
|
Chandran S, Mannon RB. T cell-mediated rejection in kidney transplant recipients: The end(point) is also the beginning. Am J Transplant 2022; 22:683-684. [PMID: 35073440 DOI: 10.1111/ajt.16964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 01/25/2023]
Affiliation(s)
- Sindhu Chandran
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, USA
| | - Roslyn B Mannon
- Division of Nephrology, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
29
|
Gowrishankar S. Banff classification from 1991 to 2019. A significant contribution to our understanding and reporting of allograft renal biopsies. Indian J Nephrol 2022; 32:1-7. [PMID: 35283563 PMCID: PMC8916159 DOI: 10.4103/ijn.ijn_270_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/04/2020] [Accepted: 10/09/2020] [Indexed: 11/05/2022] Open
Abstract
The Banff schema of classification of renal allograft biopsies, first proposed at the meeting in Banff, Canada in 1991 has evolved through subsequent meetings held once in two years and is the internationally accepted scheme of classification which is consensual, current, validated and in clinical use. This review traces the evolution of the classification and our understanding of renal transplant pathology, with emphasis on alloimmune reactions. The proceedings of the meetings and the important studies which have shaped the classification are covered.
Collapse
|
30
|
Loupy A, Mengel M, Haas M. 30 years of the International Banff Classification for Allograft Pathology: The Past, Present and Future of Kidney Transplant Diagnostics. Kidney Int 2021; 101:678-691. [DOI: 10.1016/j.kint.2021.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
|
31
|
Ravindranath MH, El Hilali F, Filippone EJ. The Impact of Inflammation on the Immune Responses to Transplantation: Tolerance or Rejection? Front Immunol 2021; 12:667834. [PMID: 34880853 PMCID: PMC8647190 DOI: 10.3389/fimmu.2021.667834] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
Transplantation (Tx) remains the optimal therapy for end-stage disease (ESD) of various solid organs. Although alloimmune events remain the leading cause of long-term allograft loss, many patients develop innate and adaptive immune responses leading to graft tolerance. The focus of this review is to provide an overview of selected aspects of the effects of inflammation on this delicate balance following solid organ transplantation. Initially, we discuss the inflammatory mediators detectable in an ESD patient. Then, the specific inflammatory mediators found post-Tx are elucidated. We examine the reciprocal relationship between donor-derived passenger leukocytes (PLs) and those of the recipient, with additional emphasis on extracellular vesicles, specifically exosomes, and we examine their role in determining the balance between tolerance and rejection. The concept of recipient antigen-presenting cell "cross-dressing" by donor exosomes is detailed. Immunological consequences of the changes undergone by cell surface antigens, including HLA molecules in donor and host immune cells activated by proinflammatory cytokines, are examined. Inflammation-mediated donor endothelial cell (EC) activation is discussed along with the effect of donor-recipient EC chimerism. Finally, as an example of a specific inflammatory mediator, a detailed analysis is provided on the dynamic role of Interleukin-6 (IL-6) and its receptor post-Tx, especially given the potential for therapeutic interdiction of this axis with monoclonal antibodies. We aim to provide a holistic as well as a reductionist perspective of the inflammation-impacted immune events that precede and follow Tx. The objective is to differentiate tolerogenic inflammation from that enhancing rejection, for potential therapeutic modifications. (Words 247).
Collapse
Affiliation(s)
- Mepur H. Ravindranath
- Department of Hematology and Oncology, Children’s Hospital, Los Angeles, CA, United States
- Terasaki Foundation Laboratory, Santa Monica, CA, United States
| | | | - Edward J. Filippone
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
32
|
Nakamura T, Shirouzu T. Antibody-Mediated Rejection and Recurrent Primary Disease: Two Main Obstacles in Abdominal Kidney, Liver, and Pancreas Transplants. J Clin Med 2021; 10:5417. [PMID: 34830699 PMCID: PMC8619797 DOI: 10.3390/jcm10225417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/08/2023] Open
Abstract
The advances in acute phase care have firmly established the practice of organ transplantation in the last several decades. Then, the next issues that loom large in the field of transplantation include antibody-mediated rejection (ABMR) and recurrent primary disease. Acute ABMR is a daunting hurdle in the performance of organ transplantation. The recent progress in desensitization and preoperative monitoring of donor-specific antibodies enables us to increase positive outcomes. However, chronic active ABMR is one of the most significant problems we currently face. On the other hand, recurrent primary disease is problematic for many recipients. Notably, some recipients, unfortunately, lost their vital organs due to this recurrence. Although some progress has been achieved in these two areas, many other factors remain largely obscure. In this review, these two topics will be discussed in light of recent discoveries.
Collapse
Affiliation(s)
- Tsukasa Nakamura
- Department of Organ Transplantation and General Surgery, Kyoto Prefectural University of Medicine, Kajii-cho 465, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takayuki Shirouzu
- Molecular Diagnositcs Division, Wakunaga Pharmaceutical Co., Ltd., 13-4 Arakicho, shinjyuku-ku, Tokyo 160-0007, Japan;
| |
Collapse
|
33
|
Moroni G, Porata G, Raffiotta F, Quaglini S, Frontini G, Sacchi L, Binda V, Calatroni M, Reggiani F, Banfi G, Ponticelli C. Beyond ISN/RPS Lupus Nephritis Classification: Adding Chronicity Index to Clinical Variables Predicts Kidney Survival. KIDNEY360 2021; 3:122-132. [PMID: 35368572 PMCID: PMC8967609 DOI: 10.34067/kid.0005512021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/01/2021] [Indexed: 01/10/2023]
Abstract
Background A renewed interest for activity and chronicity indices as predictors of lupus nephritis (LN) outcome has emerged. Revised National Institutes of Health activity and chronicity indices have been proposed to classify LN lesions, but they should be validated by future studies. The aims of this study were (1) to detect the histologic features associated with the development of kidney function impairment (KFI), and (2) to identify the best clinical-histologic model to predict KFI at time of kidney biopsy. Methods Patients with LN who had more than ten glomeruli per kidney biopsy specimen were admitted to the study. Univariate and multivariate logistic regression and Cox proportional hazards models were used to investigate whether activity and chronicity indices could predict KFI development. Results Among 203 participants with LN followed for 14 years, correlations were found between the activity index, and its components, and clinical-laboratory signs of active LN at baseline. The chronicity index was correlated with serum creatinine. Thus, serum creatinine was significantly and directly correlated with both activity and chronicity indices. In the multivariate analysis, glomerulosclerosis (OR, 3.05; 95% CI, 1.17 to 7.91; P=0.02) and fibrous crescents (OR, 6.84; 95% CI, 3.22 to 14.52; P<0.001) associated with either moderate/severe tubular atrophy (OR, 3.17; 95% CI, 1.04 to 9.64; P=0.04), or with interstitial fibrosis (OR, 2.36; 95% CI, 1.05 to 5.32; P=0.04), predicted KFI. Considering both clinical and histologic features, serum creatinine (OR, 1.68; 95% CI, 1.31 to 2.15; P<0.001), arterial hypertension (OR, 4.64; 95% CI, 1.90 to 11.32; P<0.001), glomerulosclerosis (OR, 2.12; 95% CI, 1.00 to 4.50; P=0.05), and fibrous crescents (OR, 5.18; 95% CI, 2.43 to 11.04; P<0.001) independently predicted KFI. Older age (P<0.001) and longer delay between clinical onset of LN and kidney biopsy (P<0.001) were significantly correlated with baseline chronicity index. Conclusions The chronicity index and its components, but not the activity index, were significantly associated with an impairment of kidney function. The Cox model showed that serum creatinine, arterial hypertension, chronic glomerular lesions, and delay in kidney biopsy predicted KFI. These data reinforce the importance of timely kidney biopsy in LN.
Collapse
Affiliation(s)
- Gabriella Moroni
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Giulia Porata
- Nephrology and Dialysis Unit, San Paolo Hospital, Milan, Italy
| | | | - Silvana Quaglini
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Giulia Frontini
- Nephrology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lucia Sacchi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Valentina Binda
- Nephrology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Calatroni
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | | | | |
Collapse
|
34
|
Abstract
With the incremental improvements in long-term kidney transplant survival, there is renewed focus on what causes failure of the transplanted allograft. Over the past decade, our understanding of the injuries that lead to loss of graft function over time has evolved. Chronic allograft injury includes both immune-mediated and nonimmune-mediated injuries, which may involve the organ donor, the recipient, or both. The targets of injury include the kidney tubular epithelium, the endothelium, and the glomerulus. As a response to injury, there are the expected tissue remodeling and repair processes. However, if inflammation persists, which is not uncommon in the transplant setting, the resulting maladaptive response is matrix deposition and/or fibrosis. This ultimately leads to declining graft function and, finally, failure. With our advancing knowledge of the multiple etiologies and mechanisms, enhanced by more recent cohort studies in humans, there is an opportunity to identify those at greater risk to initiate new strategies to ameliorate the process. Although the most recent studies focus on immune-mediated injuries, there is a critical need to identify both markers of injury and mechanisms of injury. In this review, we highlight the findings of recent studies, highlight the potential therapeutic targets, and identify the continued unmet need for understanding the mechanisms of late graft failure.
Collapse
Affiliation(s)
- Eric Langewisch
- Division of Nephrology, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Roslyn B. Mannon
- Division of Nephrology, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Medical Service, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
35
|
Ricaurte Archila L, Denic A, Mullan AF, Narasimhan R, Bogojevic M, Thompson RH, Leibovich BC, Sangaralingham SJ, Smith ML, Alexander MP, Rule AD. A Higher Foci Density of Interstitial Fibrosis and Tubular Atrophy Predicts Progressive CKD after a Radical Nephrectomy for Tumor. J Am Soc Nephrol 2021; 32:2623-2633. [PMID: 34531177 PMCID: PMC8722813 DOI: 10.1681/asn.2021020267] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/22/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Chronic tubulointerstitial injury on kidney biopsy is usually quantified by the percentage of cortex with interstitial fibrosis/tubular atrophy (IF/TA). Whether other patterns of IF/TA or inflammation in the tubulointerstitium have prognostic importance beyond percentage IF/TA is unclear. METHODS We obtained, stained, and digitally scanned full cortical thickness wedge sections of renal parenchyma from patients who underwent a radical nephrectomy for a tumor over 2000-2015, and morphometrically analyzed the tubulointerstitium of the cortex for percentage IF/TA, IF/TA density (foci per mm2 cortex), percentage subcapsular IF/TA, striped IF/TA, percentage inflammation (both within and outside IF/TA regions), and percentage subcapsular inflammation. Patients were followed with visits every 6-12 months. Progressive CKD was defined as dialysis, kidney transplantation, or 40% decline from the postnephrectomy eGFR. Cox models assessed the risk of CKD or noncancer mortality with morphometric measures of tubulointerstitial injury after adjustment for the percentage IF/TA and clinical characteristics. RESULTS Among 936 patients (mean age, 64 years; postnephrectomy baseline eGFR, 48 ml/min per 1.73m2), 117 progressive CKD events and 183 noncancer deaths occurred over a median 6.4 years. Higher IF/TA density predicted both progressive CKD and noncancer mortality after adjustment for percentage IF/TA and predicted progressive CKD after further adjustment for clinical characteristics. Independent of percentage IF/TA, age, and sex, higher IF/TA density correlated with lower eGFR, smaller nonsclerosed glomeruli, more global glomerulosclerosis, and smaller total cortical volume. CONCLUSIONS Higher density of IF/TA foci (a more scattered pattern with more and smaller foci) predicts higher risk of progressive CKD after radical nephrectomy compared with the same percentage of IF/TA but with fewer and larger foci.
Collapse
Affiliation(s)
| | - Aleksandar Denic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Aidan F. Mullan
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Ramya Narasimhan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Marija Bogojevic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Maxwell L. Smith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mariam P. Alexander
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Andrew D. Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Division of Epidemiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
36
|
Wei J, Chen L, Wang D, Tang L, Xie Z, Chen W, Zhang S, Weng G. Upregulation of RIP3 promotes necroptosis via a ROS‑dependent NF‑κB pathway to induce chronic inflammation in HK‑2 cells. Mol Med Rep 2021; 24:783. [PMID: 34498705 PMCID: PMC8441977 DOI: 10.3892/mmr.2021.12423] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/10/2021] [Indexed: 11/29/2022] Open
Abstract
Tubular atrophy/interstitial fibrosis (TA/IF) is a major cause of late allograft loss, and inflammation within areas of TA/IF is associated with adverse outcomes in kidney transplantation. However, there is currently no satisfactory method to suppress this inflammation to improve TA/IF. The present study aimed to determine the proinflammatory role of receptor-interacting protein 3 (RIP3) in TA/IF to discover a novel therapeutic target. Reverse transcription-quantitative PCR and western blotting were performed to detect the expression of RIP3 and inflammation-associated factors. Lactate dehydrogenase release assay was used to determine necroptosis. Fluorescent 2,7-dichlorodihydrofluorescein diacetate was used to detect the levels of reactive oxygen species (ROS). The results demonstrated that patients with chronic TA/IF exhibited upregulated receptor-interacting protein 3 (RIP3) expression compared with the patients who had a favorable recovery after renal transplant. Therefore, the current study used normal renal tubular epithelial cells HK-2 to establish a cellular model with a high expression level of RIP3 in order to investigate the effect of RIP3 on renal epithelial cells after transplantation. The western blotting results demonstrated that overexpression of RIP3 could significantly increase the phosphorylation level of the necroptosis executive molecule mixed lineage kinase domain-like protein. Lactate dehydrogenase release, a key feature of necroptosis, was also markedly improved by RIP3 overexpression. Moreover, a higher inflammatory response was detected in HK-2 cells with RIP3 overexpression, and this elevated inflammation could be restored by the necroptosis inhibitor necrosulfonamide. Of note, it was found that overexpression of RIP3 activated the NF-κB signaling pathway via the excessive accumulation of ROS to induce necroptosis, which ultimately led to inflammation. Collectively, these findings indicated that overexpression of RIP3 promoted necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation, suggesting that RIP3 may have the potential to be a therapeutic target against inflammation in TA/IF.
Collapse
Affiliation(s)
- Junjun Wei
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Liangliang Chen
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Duidui Wang
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Li Tang
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Zhenhua Xie
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Weifeng Chen
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Shuwei Zhang
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Guobin Weng
- Department of Renal Transplantation, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315100, P.R. China
| |
Collapse
|
37
|
Handschin J, Wehmeier C, Amico P, Hopfer H, Dickenmann M, Schaub S, Hirt-Minkowski P. Urinary CXCL10 Measurement in Late Renal Allograft Biopsies Predicts Outcome Even in Histologically Quiescent Patients. Transplant Proc 2021; 53:2168-2179. [PMID: 34419254 DOI: 10.1016/j.transproceed.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/27/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND CXCL10 is a promising early noninvasive diagnostic marker for allograft rejection and predictive for long-term outcomes. However, its value when measured later in the posttransplant course has not yet been accurately analyzed. METHODS We investigated urinary CXCL10 in 141 patients from a prospective, observational renal transplant cohort with 182 clinically indicated allograft biopsies performed >12 months posttransplant and corresponding urines. Urinary CXCL10 was retrospectively quantified on stored urines using the MSD V-Plex Chemokine Panel 1 sandwich immunoassay (Meso Scale Discovery). The primary outcome was a composite of allograft loss/renal function decline (>30% estimated glomerular filtration rate [eGFR]-decrease between index biopsy and last follow-up). RESULTS Seventy-two patients (51%) reached the primary outcome, and their urinary CXCL10 levels were significantly higher at the time of their biopsy compared with patients with stable allograft function (median 9.3 ng/mmol vs 3.3 ng/mmol, P < .0001). Time-to-endpoint analyses according to high/low urinary CXCL10 demonstrated that low urinary CXCL10 (≤7.0 ng/mmol) was associated with 73% 5-year event-free graft survival compared with 48% with high urinary CXCL10 (>7.0 ng/mmol; P = .0001). Even in histologically quiescent patients, high urinary CXCL10 was associated with inferior endpoint-free graft survival (P = .003), and it was an independent predictor of the primary outcome (P = .03). CONCLUSIONS This study demonstrates that urinary CXCL10 has a promising diagnostic performance for detection of late allograft rejection and is an independent predictor of long-term renal allograft outcomes, even in histologically quiescent patients.
Collapse
Affiliation(s)
- Joelle Handschin
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Caroline Wehmeier
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Patrizia Amico
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Helmut Hopfer
- lnstitute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Michael Dickenmann
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Stefan Schaub
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland; Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland; HLA-Diagnostic and lmmunogenetics, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Patricia Hirt-Minkowski
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
38
|
Kowalewska J, El Moudden I, Perkowska-Ptasinska A, Kapp ME, Fogo AB, Lin MY, Mirza A, Troyer DA, Durlik M, Sandhu R, Ciszek M, Deborska-Materkowska D, Kuczynski D, McCune TR. Assessment of the Banff Working Group classification of definitive BK polyomavirus nephropathy. Transpl Int 2021; 34:2286-2296. [PMID: 34339576 DOI: 10.1111/tri.14003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022]
Abstract
Polyomavirus associated nephropathy (PyVAN) continues to be a burden in renal transplantation leading to allograft insufficiency or graft failure. A presumptive diagnosis of PyVAN is made based on the presence of BK polyomavirus in patients' plasma; however, kidney biopsy remains the gold standard to establish a definitive diagnosis. The Banff Working Group on PyVAN proposed a novel classification of definitive PyVAN based on polyomavirus replication/load level and the extent of interstitial fibrosis. The aim of our study was to test the newly defined classes of PyVAN using independent cohorts of 124 kidney transplant patients with PyVAN with respect to the initial presentation and outcome, and to compare our analysis to that previously reported. Detailed analysis of our cohort revealed that the proposed classification of PyVAN did not stratify or identify patients at increased risk of allograft failure. Specifically, while class 3 was associated with the worst prognosis, there was no significant difference between the outcomes in classes 1 and 2. We also found that the timing post-transplantation and inflammation in areas of interstitial fibrosis and tubular atrophy might be additional factors contributing to an unfavorable allograft outcome in patients with PyVAN.
Collapse
Affiliation(s)
- Jolanta Kowalewska
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ismail El Moudden
- EVMS-Sentara Healthcare Analytics and Delivery Science Institute, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | - Meghan E Kapp
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mercury Y Lin
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Alamgir Mirza
- Department of Medicine, Division of Nephrology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Dean A Troyer
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Magdalena Durlik
- Department of Transplantation Medicine and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Rana Sandhu
- Cedars Sinai Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Michal Ciszek
- Department of Transplantation Medicine and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | | | - Daniel Kuczynski
- Department of Transplantation Medicine and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Thomas R McCune
- Department of Medicine, Division of Nephrology, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
39
|
Kung VL, Sandhu R, Haas M, Huang E. Chronic active T cell–mediated rejection is variably responsive to immunosuppressive therapy. Kidney Int 2021; 100:391-400. [DOI: 10.1016/j.kint.2021.03.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/24/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
|
40
|
Garcia-Sanchez C, Casillas-Abundis MA, Pinelli DF, Tambur AR, Hod-Dvorai R. Impact of SIRPα polymorphism on transplant outcomes in HLA-identical living donor kidney transplantation. Clin Transplant 2021; 35:e14406. [PMID: 34180101 DOI: 10.1111/ctr.14406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022]
Abstract
Signal-regulatory protein α (SIRPα), a polymorphic inhibitory membrane-bound receptor, and its ligand CD47 have recently been implicated in the modulation of innate immune allorecognition in murine models. Here, we investigate the potential impact of SIRPα donor-recipient mismatches on graft outcomes in human kidney transplantation. To eliminate the specific role of HLA-matching in alloresponse, we genotyped the two most common variants of SIRPα in a cohort of 55 HLA-identical, biologically-related, donor-recipient pairs. 69% of pairs were SIRPα identical. No significant differences were found between donor-recipient SIRPα-mismatch status and T cell-mediated rejection/borderline changes (25.8% vs. 25%) or slow graft function (15.8% vs. 17.6%). A trend towards more graft failure (GF) (23.5% vs. 5.3%, P = .06), interstitial inflammation (50% vs. 23%, P = .06) and significant changes in peritubular capillaritis (ptc) (25% vs. 0%, P = .02) were observed in the SIRPα-mismatched group. Unexpectedly, graft-versus-host (GVH) SIRPα-mismatched pairs exhibited higher rates of GF and tubulitis (38% vs. 5%, P = .031 and .61 ± .88 vs. 0, P = .019; respectively). Whether the higher prevalence of ptc in SIRPα-mismatched recipients and the higher rates of GF in GVH SIRPα-mismatched pairs represent a potential role for SIRPα in linking innate immunity and alloimmune rejection requires further investigation in larger cohorts.
Collapse
Affiliation(s)
- Cynthia Garcia-Sanchez
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - M Aurora Casillas-Abundis
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - David F Pinelli
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Anat R Tambur
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Reut Hod-Dvorai
- Pathology Department, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
41
|
Hart A, Schladt DP, Matas AJ, Itzler R, Israni AK, Kasiske BL. Incidence, risk factors, and long-term outcomes associated with antibody-mediated rejection - The long-term Deterioration of Kidney Allograft Function (DeKAF) prospective cohort study. Clin Transplant 2021; 35:e14337. [PMID: 33955070 DOI: 10.1111/ctr.14337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023]
Abstract
Major gaps remain in our understanding of antibody-mediated rejection (AMR) after kidney transplant. We examined the incidence, risk factors, response to treatment, and effects on outcomes of AMR at seven transplant programs in the long-term Deterioration of Kidney Allograft Function prospective study cohort. Among 3131 kidney recipients, there were 194 observed AMR cases (6.2%) during (mean ± SD) 4.85 ± 1.86 years of follow-up. Time to AMR was 0.97 ± 1.17 (median, 0.48) years. Risk factors for AMR included younger recipient age, human leukocyte antigen DR mismatches, panel-reactive antibody >0%, positive T- or B-cell cross-match, and delayed graft function. Compared with no AMR, the adjusted time-dependent hazard ratio for death-censored graft failure is 10.1 (95% confidence interval, 6.5-15.7) for all AMR patients, 4.0 (2.5, 9.1) for early AMR (<90 days after transplant), and 24.0 (14.0-41.1) for late AMR (≥90 days after transplant). Patients were treated with different therapeutic combinations. Of 194 kidney transplant recipients with AMR, 50 (25.8%) did not respond to treatment, defined as second AMR within 100 days or no improvement in estimated glomerular filtration rate by 42 days. Long-term outcomes after AMR are poor, regardless of the initial response to treatment. Better prevention and new therapeutic strategies are needed to improve long-term allograft survival.
Collapse
Affiliation(s)
- Allyson Hart
- Department of Medicine, Hennepin County Medical Center, Hennepin Healthcare, Minneapolis, MN, USA.,Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | - David P Schladt
- Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | - Arthur J Matas
- Transplantation Division, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - Ajay K Israni
- Department of Medicine, Hennepin County Medical Center, Hennepin Healthcare, Minneapolis, MN, USA.,Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | - Bertram L Kasiske
- Department of Medicine, Hennepin County Medical Center, Hennepin Healthcare, Minneapolis, MN, USA.,Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| |
Collapse
|
42
|
Clinical Relevance of Corticosteroid Withdrawal on Graft Histological Lesions in Low-Immunological-Risk Kidney Transplant Patients. J Clin Med 2021; 10:jcm10092005. [PMID: 34067039 PMCID: PMC8125434 DOI: 10.3390/jcm10092005] [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: 03/19/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
The impact of corticosteroid withdrawal on medium-term graft histological changes in kidney transplant (KT) recipients under standard immunosuppression is uncertain. As part of an open-label, multicenter, prospective, phase IV, 24-month clinical trial (ClinicalTrials.gov, NCT02284464) in low-immunological-risk KT recipients, 105 patients were randomized, after a protocol-biopsy at 3 months, to corticosteroid continuation (CSC, n = 52) or corticosteroid withdrawal (CSW, n = 53). Both groups received tacrolimus and MMF and had another protocol-biopsy at 24 months. The acute rejection rate, including subclinical inflammation (SCI), was comparable between groups (21.2 vs. 24.5%). No patients developed dnDSA. Inflammatory and chronicity scores increased from 3 to 24 months in patients with, at baseline, no inflammation (NI) or SCI, regardless of treatment. CSW patients with SCI at 3 months had a significantly increased chronicity score at 24 months. HbA1c levels were lower in CSW patients (6.4 ± 1.2 vs. 5.7 ± 0.6%; p = 0.013) at 24 months, as was systolic blood pressure (134.2 ± 14.9 vs. 125.7 ± 15.3 mmHg; p = 0.016). Allograft function was comparable between groups and no patients died or lost their graft. An increase in chronicity scores at 2-years post-transplantation was observed in low-immunological-risk KT recipients with initial NI or SCI, but CSW may accelerate chronicity changes, especially in patients with early SCI. This strategy did, however, improve the cardiovascular profiles of patients.
Collapse
|
43
|
Helgeson ES, Mannon R, Grande J, Gaston RS, Cecka MJ, Kasiske BL, Rush D, Gourishankar S, Cosio F, Hunsicker L, Connett J, Matas AJ. i-IFTA and chronic active T cell-mediated rejection: A tale of 2 (DeKAF) cohorts. Am J Transplant 2021; 21:1866-1877. [PMID: 33052625 DOI: 10.1111/ajt.16352] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 01/25/2023]
Abstract
Inflammation in areas of fibrosis (i-IFTA) in posttransplant biopsies is part of the diagnostic criteria for chronic active TCMR (CA TCMR -- i-IFTA ≥ 2, ti ≥ 2, t ≥ 2). We evaluated i-IFTA and CA TCMR in the DeKAF indication biopsy cohorts: prospective (n = 585, mean time to biopsy = 1.7 years); cross-sectional (n = 458, mean time to biopsy = 7.8 years). Grouped by i-IFTA scores, the 3-year postbiopsy DC-GS is similar across cohorts. Although a previous acute rejection episode (AR) was more common in those with i-IFTA on biopsy, the majority of those with i-IFTA had not had previous AR. There was no association between type of previous AR (AMR, TCMR) and presence of i-IFTA. In both cohorts, i-IFTA was associated with markers of both cellular (increased Banff i, t, ti) and humoral (increased g, ptc, C4d, DSA) activity. Biopsies with i-IFTA = 1 and i-IFTA ≥ 2 with concurrent t ≥ 2 and ti ≥ 2 had similar DC-GS. These results suggest that (a) i-IFTA≥1 should be considered a threshold for diagnoses incorporating i-IFTA, ti, and t; (b) given that i-IFTA ≥ 2,t ≥ 2, ti ≥ 2 can occur in the absence of preceding TCMR and that the component histologic scores (i-IFTA,t,ti) each indicate an acute change (albeit i-IFTA on the nonspecific background of IFTA), the diagnostic category "CA TCMR" should be reconsidered.
Collapse
Affiliation(s)
- Erika S Helgeson
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Roslyn Mannon
- University of Nebraska Medical Center and Nebraska-Western Iowa Veterans Affairs Medical Center, Omaha, Nebraska
| | | | - Robert S Gaston
- University of Nebraska Medical Center and Nebraska-Western Iowa Veterans Affairs Medical Center, Omaha, Nebraska
| | - Michael J Cecka
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | | | - David Rush
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sita Gourishankar
- Department of Medicine, Division of Nephrology, Univeristy of Alberta, Edmonton, Alberta, Canada
| | | | | | - John Connett
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Arthur J Matas
- Department of Surgery, Transplantation Division, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
44
|
Paul RS, Almokayad I, Collins A, Raj D, Jagadeesan M. Donor-derived Cell-free DNA: Advancing a Novel Assay to New Heights in Renal Transplantation. Transplant Direct 2021; 7:e664. [PMID: 33564715 PMCID: PMC7862009 DOI: 10.1097/txd.0000000000001098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Despite advances in transplant immunosuppression, long-term renal allograft outcomes remain suboptimal because of the occurrence of rejection, recurrent disease, and interstitial fibrosis with tubular atrophy. This is largely due to limitations in our understanding of allogeneic processes coupled with inadequate surveillance strategies. The concept of donor-derived cell-free DNA as a signal of allograft stress has therefore rapidly been adopted as a noninvasive monitoring tool. Refining it for effective clinical use, however, remains an ongoing effort. Furthermore, its potential to unravel new insights in alloimmunity through novel molecular techniques is yet to be realized. This review herein summarizes current knowledge and active endeavors to optimize cell-free DNA-based diagnostic techniques for clinical use in kidney transplantation. In addition, the integration of DNA methylation and microRNA may unveil new epigenetic signatures of allograft health and is also explored in this report. Directing research initiatives toward these aspirations will not only improve diagnostic precision but may foster new paradigms in transplant immunobiology.
Collapse
Affiliation(s)
- Rohan S. Paul
- Division of Kidney Disease & Hypertension, George Washington University, Washington, DC
| | - Ismail Almokayad
- Division of Kidney Disease & Hypertension, George Washington University, Washington, DC
| | - Ashte Collins
- Division of Kidney Disease & Hypertension, George Washington University, Washington, DC
| | - Dominic Raj
- Division of Kidney Disease & Hypertension, George Washington University, Washington, DC
| | | |
Collapse
|
45
|
Jahn L, Rüster C, Schlosser M, Winkler Y, Foller S, Grimm MO, Wolf G, Busch M. Rate, Factors, and Outcome of Delayed Graft Function After Kidney Transplantation of Deceased Donors. Transplant Proc 2021; 53:1454-1461. [PMID: 33612277 DOI: 10.1016/j.transproceed.2021.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/08/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Delayed graft function (DGF) is a frequent complication after kidney transplantation affecting long-term outcome. PATIENTS AND METHODS A total of 525 consecutive recipients (age 54.2 ± 13.4 years, 33% female) of kidneys from deceased donors transplanted between 2005 and 2012 were retrospectively examined. DGF was defined as the need of dialysis within the first week after transplantation. RESULTS DGF developed in 21.1% (n = 111). Factors associated with DGF (P ≤ .035, respectively) were recipient body mass index, C-reactive protein of the recipient, residual diuresis, cold ischemia time, donor age, and diuresis in the first hour after transplantation. Median duration of DGF was 16 (2-66) days. Patients after DGF had a significantly lower GFR compared with recipients without DGF either after 3 (32.9 ± 16.5 vs 46.3 ± 18.4 mL/min/1.73 m2) or after 12 months (38.9 ± 19.3 vs 48.6 ± 20.4 mL/min/1.73 m2, P < .001, resp.). During DGF, 12.4% developed BANFF II and 18.0% BANFF I rejection, 20.2% had signs of transplant glomerulitis (first biopsy), and 16.2% (n = 18) remained on dialysis. CONCLUSION DGF affects 1 out of 5 kidney transplants from deceased donors. Minimizing modifiable risk factors, in particular immunologic risk, may ameliorate the incidence and outcome of DGF. The outcome of DGF depends mainly on the diagnosis of any rejection and worsens upon detection of transplant glomerulitis and pronounced interstitial fibrosis and tubular atrophy (IFTA).
Collapse
Affiliation(s)
- Laura Jahn
- Department of Internal Medicine III/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Christiane Rüster
- Department of Internal Medicine III/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Mandy Schlosser
- Department of Internal Medicine III/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Yvonne Winkler
- Department of Urology/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Susan Foller
- Department of Urology/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Marc-Oliver Grimm
- Department of Urology/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany
| | - Martin Busch
- Department of Internal Medicine III/Collaborative Kidney Transplant Center, University Hospital Jena - Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
46
|
Molecular Analysis of Renal Allograft Biopsies: Where Do We Stand and Where Are We Going? Transplantation 2021; 104:2478-2486. [PMID: 32150035 DOI: 10.1097/tp.0000000000003220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A renal core biopsy for histological evaluation is the gold standard for diagnosing renal transplant pathology. However, renal biopsy interpretation is subjective and can render insufficient precision, making it difficult to apply a targeted therapeutic regimen for the individual patient. This warrants a need for additional methods assessing disease state in the renal transplant. Significant research activity has been focused on the role of molecular analysis in the diagnosis of renal allograft rejection. The identification of specific molecular expression patterns in allograft biopsies related to different types of allograft injury could provide valuable information about the processes underlying renal transplant dysfunction and can be used for the development of molecular classifier scores, which could improve our diagnostic and prognostic ability and could guide treatment. Molecular profiling has the potential to be more precise and objective than histological evaluation and may identify injury even before it becomes visible on histology, making it possible to start treatment at the earliest time possible. Combining conventional diagnostics (histology, serology, and clinical data) and molecular evaluation will most likely offer the best diagnostic approach. We believe that the use of state-of-the-art molecular analysis will have a significant impact in diagnostics after renal transplantation. In this review, we elaborate on the molecular phenotype of both acute and chronic T cell-mediated rejection and antibody-mediated rejection and discuss the additive value of molecular profiling in the setting of diagnosing renal allograft rejection and how this will improve transplant patient care.
Collapse
|
47
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
48
|
Impact of Subclinical Borderline Inflammation on Kidney Transplant Outcomes. Transplant Direct 2021; 7:e663. [PMID: 33511268 PMCID: PMC7837932 DOI: 10.1097/txd.0000000000001119] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. Surveillance biopsies permit early detection of subclinical inflammation before clinical dysfunction, but the impact of detecting early subclinical phenotypes remains unclear. Methods. We conducted a single-center retrospective cohort study of 441 consecutive kidney transplant recipients between 2015 and 2018 with surveillance biopsies at 6 months post-transplant. We tested the hypothesis that early subclinical inflammation (subclinical borderline changes, T cell-mediated rejection, or microvascular injury) is associated with increased incidence of a composite endpoint including acute rejection and allograft failure. Results. Using contemporaneous Banff criteria, we detected subclinical inflammation in 31%, with the majority (75%) having a subclinical borderline phenotype (at least minimal inflammation with mild tubulitis [>i0t1]). Overall, subclinical inflammation was independently associated with the composite endpoint (adjusted hazard ratio, 2.88; 1.11-7.51; P = 0.03). The subgroup with subclinical borderline inflammation, predominantly those meeting the Banff 2019 i1t1 threshold, was independently associated with 5-fold increased hazard for the composite endpoint (P = 0.02). Those with concurrent subclinical inflammation and subclinical chronic allograft injury had worse outcomes. The effect of treating subclinical inflammation was difficult to ascertain in small heterogeneous subgroups. Conclusions. Subclinical acute and chronic inflammation are common at 6 months post-transplant in kidney recipients with stable allograft function. The subclinical borderline phenotype with both tubulitis and interstitial inflammation was independently associated with poor long-term outcomes. Further studies are needed to elucidate the role of surveillance biopsies for management of allograft inflammation in kidney transplantation.
Collapse
|
49
|
Chamoun B, Torres IB, Gabaldón A, Sellarés J, Perelló M, Castellá E, Guri X, Salcedo M, Toapanta NG, Cidraque I, Moreso F, Seron D. Progression of Interstitial Fibrosis and Tubular Atrophy in Low Immunological Risk Renal Transplants Monitored by Sequential Surveillance Biopsies: The Influence of TAC Exposure and Metabolism. J Clin Med 2021; 10:jcm10010141. [PMID: 33406589 PMCID: PMC7796060 DOI: 10.3390/jcm10010141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
The combination of tacrolimus (TAC) and mycophenolate is the most widely employed maintenance immunosuppression in renal transplants. Different surrogates of tacrolimus exposure or metabolism such as tacrolimus trough levels (TAC-C0), coefficient of variation of tacrolimus (CV-TAC-C0), time in therapeutic range (TTR), and tacrolimus concentration dose ratio (C/D) have been associated with graft outcomes. We explore in a cohort of low immunological risk renal transplants (n = 85) treated with TAC, mycophenolate mofetil (MMF), and steroids and then monitored by paired surveillance biopsies the association between histological lesions and TAC-C0 at the time of biopsy as well as CV-TAC-C0, TTR, and C/D during follow up. Interstitial inflammation (i-Banff score ≥ 1) in the first surveillance biopsy was associated with TAC-C0 (odds ratio (OR): 0.69, 95% confidence interval (CI): 0.50–0.96; p = 0.027). In the second surveillance biopsy, inflammation was associated with time below the therapeutic range (OR: 1.05 and 95% CI: 1.01–1.10; p = 0.023). Interstitial inflammation in scarred areas (i-IFTA score ≥ 1) was not associated with surrogates of TAC exposure/metabolism. Progression of interstitial fibrosis/tubular atrophy (IF/TA) was observed in 35 cases (41.2%). Multivariate regression logistic analysis showed that mean C/D (OR: 0.48; 95% CI: 0.25–0.92; p = 0.026) and IF/TA in the first biopsy (OR: 0.43, 95% CI: 0.24–0.77, p = 0.005) were associated with IF/TA progression between biopsies. A low C/D ratio is associated with IF/TA progression, suggesting that TAC nephrotoxicity may contribute to fibrosis progression in well immunosuppressed patients. Our data support that TAC exposure is associated with inflammation in healthy kidney areas but not in scarred tissue.
Collapse
Affiliation(s)
- Betty Chamoun
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Irina B. Torres
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Alejandra Gabaldón
- Pathology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (A.G.); (M.S.)
| | - Joana Sellarés
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Manel Perelló
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Eva Castellá
- Radiology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (E.C.); (X.G.)
| | - Xavier Guri
- Radiology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (E.C.); (X.G.)
| | - Maite Salcedo
- Pathology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (A.G.); (M.S.)
| | - Nestor G. Toapanta
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Ignacio Cidraque
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
| | - Francesc Moreso
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
- Department of Medicine, Autonomous University of Barcelona, 08035 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-274-46-66
| | - Daniel Seron
- Nephrology Departments, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (B.C.); (I.B.T.); (J.S.); (M.P.); (N.G.T.); (I.C.); (D.S.)
- Department of Medicine, Autonomous University of Barcelona, 08035 Barcelona, Spain
| |
Collapse
|
50
|
Nakagawa K, Tsuchimoto A, Ueki K, Matsukuma Y, Okabe Y, Masutani K, Unagami K, Kakuta Y, Okumi M, Nakamura M, Nakano T, Tanabe K, Kitazono T. Significance of revised criteria for chronic active T cell-mediated rejection in the 2017 Banff classification: Surveillance by 1-year protocol biopsies for kidney transplantation. Am J Transplant 2021; 21:174-185. [PMID: 32484280 DOI: 10.1111/ajt.16093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/25/2023]
Abstract
Diagnostic criteria for chronic active T cell-mediated rejection (CA-TCMR) were revised in the Banff 2017 consensus, but it is unknown whether the new criteria predict graft prognosis of kidney transplantation. We enrolled 406 kidney allograft recipients who underwent a 1-year protocol biopsy (PB) and investigated the diagnostic significance of Banff 2017. Interobserver reproducibility of the 3 diagnosticians showed a substantial agreement rate of 0.68 in Fleiss's kappa coefficient. Thirty-three patients (8%) were classified as CA-TCMR according to Banff 2017, and 6 were previously diagnosed as normal, 12 as acute TCMR, 10 with borderline changes, and 5 as CA-TCMR according to Banff 2015 criteria. Determinant factors of CA-TCMR were cyclosporine use (vs tacrolimus), previous acute rejection, and BK polyomavirus-associated nephropathy. In survival analysis, the new diagnosis of CA-TCMR predicted a composite graft endpoint defined as doubling serum creatinine or death-censored graft loss (log-rank test, P < .001). In multivariate analysis, CA-TCMR was associated with the second highest risk of the composite endpoint (hazard ratio: 5.42; 95% confidence interval, 2.02-14.61; P < .001 vs normal) behind antibody-mediated rejection. In conclusion, diagnosis of CA-TCMR in Banff 2017 may facilitate detecting an unfavorable prognosis of kidney allograft recipients who undergo a 1-year PB.
Collapse
Affiliation(s)
- Kaneyasu Nakagawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiro Tsuchimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Ueki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuta Matsukuma
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuhiro Okabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Masutani
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kohei Unagami
- Department of Organ Transplant Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoichi Kakuta
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayoshi Okumi
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|